57
Viral vectors

Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Embed Size (px)

Citation preview

Page 1: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Viralvectors

ViralvectorsareusedforthedeliveryofgeneticalmaterialintocellsBecauseviruseshaveanaturalabilitytoefficientlydelivertheir genome into host cells they are an ideal tool for genetransfer and are often used by molecular biologists to delivertherapeuticgenes

TransductionAbortive(non-replicative)infectionthatintroducesfunctionalgeneticinformationexpressedfromtherecombinantvectorsintothetargetcell

bull Gene therapy (deliver a gene to patientswho lack the gene or carry defectivevertions)

bull Todeliveyantigens(viralvaccines)bull Viraloncotherapybull Researchuses

Virus vectors

Viral Vectors

bull Non-enveloped virus bull The capsid diameter is about 60-90 nm bull Icosahedral symmetry with projections in each of the of the 5 fold axis

AdenovirusAdenovirusampvectors

copyPrinciplesofVirologyASMPress

bull Linear double-stranded DNA (non-segmented) of 35-36kb bull Genes are codified in both strands with overlapping transcription units bull The genome has a) terminally redundant sequences which have inverted terminal repetitions (ITR)agrave origin of replication b) Packaging sequence (ψ)

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 2: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

ViralvectorsareusedforthedeliveryofgeneticalmaterialintocellsBecauseviruseshaveanaturalabilitytoefficientlydelivertheir genome into host cells they are an ideal tool for genetransfer and are often used by molecular biologists to delivertherapeuticgenes

TransductionAbortive(non-replicative)infectionthatintroducesfunctionalgeneticinformationexpressedfromtherecombinantvectorsintothetargetcell

bull Gene therapy (deliver a gene to patientswho lack the gene or carry defectivevertions)

bull Todeliveyantigens(viralvaccines)bull Viraloncotherapybull Researchuses

Virus vectors

Viral Vectors

bull Non-enveloped virus bull The capsid diameter is about 60-90 nm bull Icosahedral symmetry with projections in each of the of the 5 fold axis

AdenovirusAdenovirusampvectors

copyPrinciplesofVirologyASMPress

bull Linear double-stranded DNA (non-segmented) of 35-36kb bull Genes are codified in both strands with overlapping transcription units bull The genome has a) terminally redundant sequences which have inverted terminal repetitions (ITR)agrave origin of replication b) Packaging sequence (ψ)

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 3: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

bull Gene therapy (deliver a gene to patientswho lack the gene or carry defectivevertions)

bull Todeliveyantigens(viralvaccines)bull Viraloncotherapybull Researchuses

Virus vectors

Viral Vectors

bull Non-enveloped virus bull The capsid diameter is about 60-90 nm bull Icosahedral symmetry with projections in each of the of the 5 fold axis

AdenovirusAdenovirusampvectors

copyPrinciplesofVirologyASMPress

bull Linear double-stranded DNA (non-segmented) of 35-36kb bull Genes are codified in both strands with overlapping transcription units bull The genome has a) terminally redundant sequences which have inverted terminal repetitions (ITR)agrave origin of replication b) Packaging sequence (ψ)

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 4: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Viral Vectors

bull Non-enveloped virus bull The capsid diameter is about 60-90 nm bull Icosahedral symmetry with projections in each of the of the 5 fold axis

AdenovirusAdenovirusampvectors

copyPrinciplesofVirologyASMPress

bull Linear double-stranded DNA (non-segmented) of 35-36kb bull Genes are codified in both strands with overlapping transcription units bull The genome has a) terminally redundant sequences which have inverted terminal repetitions (ITR)agrave origin of replication b) Packaging sequence (ψ)

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 5: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

bull Non-enveloped virus bull The capsid diameter is about 60-90 nm bull Icosahedral symmetry with projections in each of the of the 5 fold axis

AdenovirusAdenovirusampvectors

copyPrinciplesofVirologyASMPress

bull Linear double-stranded DNA (non-segmented) of 35-36kb bull Genes are codified in both strands with overlapping transcription units bull The genome has a) terminally redundant sequences which have inverted terminal repetitions (ITR)agrave origin of replication b) Packaging sequence (ψ)

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 6: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

bull Linear double-stranded DNA (non-segmented) of 35-36kb bull Genes are codified in both strands with overlapping transcription units bull The genome has a) terminally redundant sequences which have inverted terminal repetitions (ITR)agrave origin of replication b) Packaging sequence (ψ)

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 7: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Genome functional organization

E1A E1BE2AE2BE3E4

OnsetDNAsynthesis

Inhibits apoptosis(darrp53) Block host mRNA transport Stimulate viral mRNA transport

Transctivates all other early genes and stimulates the host cell to enter an S phase-like state

Viral DNA replication

Early Late

Involved in modulating the IR of infected cells

Structural proteins

Facilitate DNA replication enhance late gene expression and decrease host protein synthesis

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 8: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Adenovirusampvectors

bull EfficientlyinfectpostmitoMccells

bull Fast(48h)onsetofgeneexpression

bull EpisomalminimalriskofinserMonmutagenesis

bull Upto37kbcapacity

bull PureconcentratedprepsrouMne

bull gt50humanserotypesanimalserotypes

bull Drawbackimmunity

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 9: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

gt Presence of E1-like factors in many cellscan allow for expression of viral proteinswhich normally initiate a strong immuneresponse dampening the efficacy of a firstgenerationgtSpace

gtReducedyieldsgtAlthoughattenuatedtoxicity towards thevectorscan be seen stable transgene expression isgenerallynotseen

Adenoviralvectors

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 10: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

These vectors whenused in vivo result inlong term high-leveltransgene expressionc o m b i n e d w i t hnegligibletoxicity

The main disadvantage for these vectors is the complexproductionduetotheneednotonlyforasuitableproducercell line but also a complementing Ad vector (helper Advector _ HAd) which provides the necessary packagingproteinsoftheviralparticle

The requirement of the HAd brings about the problem ofcontaminationfromtheHAdwhenusingE1complementingproducercells

Adenovirus vectors

insert

bull Third generation vectors all genes deleted contain only two ITRs and psi

bull Require helper virus which is E1-deleted

copyPrinciples of Virology ASM Press

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 11: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

11

FirstgenerationAdvectorsproduction

AdEasy Adenoviral Vector System 5

FIGURE 1 Production of recombinant adenovirus

gene of interest

Ori

Ori

Kan

Pac I

Pac I

Pac I

RecombinantAd

Plasmid

Transfect AD-293 (or HEK293) cells

Digest with IPac

LITR

Encapsidation signal

RITR

LITREncapsidation

signal

Promotergene of interest

poly AAdenoviral DNA

Pac IPac I

cotransformB 5183 cells select for Kan

JR

linearize with IPme

MCSShuttle Vector

R ight A

rm

Ori

Kan

gene of interest

Pac I

Pac I

LITREncapsidation

signal

RIT

R

poly APme ILeft ArmCloning Geneof Interest

HomologousRecombination

in Bacteriain vivo

Virus Productionin AD-293 Cells

Regions ofHomologous

Recombination

Left Arm

pAdEasy-1Vector

Ori Right Arm

Pac I

Amp

RITR

RIT

R

RIT

R

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 12: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

HighcapacityAdvectorsproduction

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 13: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

13

bull  It is a site-specific recombination strategy that can be used for inversion deletion translocation and insertion of DNA sequence

bull  Cre-Lox recombination system consists of Cre recombinase enzyme Sequence called Lox P site or simply Lox sequence (on which Cre recombinase acts) Natural Function of Cre-Lox System The system was initially discovered in a temperate bacteriophage P1 Functions of Cre-Lox system in P1 bacteriophage are bull  To circularise viral DNA once it is injected into host cell Hence P1 DNA exists as plasmid inside the host

instead of integrating into host genome to form prophage bull  During division to separate interlinked circularised viral DNAs so that they are passed equally to daughter

cells bull  Copy number maintenance

Lox P Site Lox P site is a 34 bp sequence which consists of 8 bp asymmetric core sequence (it determines the directionality of the Lox P site) 13 bp flanking inverted repeats Lox P sequence representing the asymmetric core and the flanking inverted repeat Cre Recombinase Cre recombinase is the product of Cyclization recombination gene (38 Kda) One Cre recombinase binds to single inverted repeat (ie on a single Lox sequence two Cre molecule binds)

bull  Cre recombinase and Lox sequence are not native to plants and animals

Cre-Lox P system

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 14: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

VectorProduction

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 15: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

FirstgenerationvectorproductionHEK293 agrave Is the major host cell line for Ad vector development including development of master cell banks and development and optimization of Ad-manufacturing processes agrave Is problematic for large-scale viral production andclinicalapplications(1RCAin3x1010viralparticles)

PerC6agraveBest design and master cell banks for GMP Adproductionagravehas strict licensing costsuses preventing manylaboratoriesfromworkingwiththiscelllineagrave is considered less robust thanHEK293 cell lineasitislessadaptabletoserumfreecultureforlargescaleproductionAdvectors

VLI-293InsertionofspacerDNA(8kb)(followingnt3510)agrave this does not prevent homologous recombinationduetoretentionofhomologybetweenthegenomeandAdvectorbutpackagingoftheAdvectorwillbesuppressedduetotheincreasedsize

HeLaisnotallowedforcommercialusebecauseofitshightumorigenicity

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 16: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Secondgenerationvectorproduction

One of the main differences from E1 complementing cell lines where E1A and E1B are expressed constitutively is that several of these cell lines rely on the use of conditionally active transcription units or gene products to control the expression of these proteins as the viral products from E2A and E4 regions are toxic to the cells

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 17: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

It should be noted that evenwith thistype of system contamination is notfully eliminated (01 to 1 is usuallyseen) but physical separation of theHC -Ad and HAd t h rough C sC lultracentrifugation can address thisissueFor small preparations this is generally acceptable however forindustrial-scaleclinicalgradevectorproductionfurthermeasureshave to be taken especially as some of this contamination cancontainpackagingcompetentHAd

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 18: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

T h i s i s a m a j o rconcernwhen vectorsare use in humantrials

Pre-existing immunity

Marked liver tropism

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 19: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

19

SetbackampJesseampGelsinger

bull Firstpersontodieinagenetherapyclinicaltrial

bull Ornithinetranscarbamylasedeficiency

bull GivenAdvectorwithnormalOTCgeneatUPenn

bull Died4dayslatermassiveimmuneresponsemulMpleorganfailure

bull Severalrulesofconductbroken

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 20: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

ImmuneresponsetoAdvectors80 of the human populationhas been exposed to at leastone human Ad serotype andhas developed a serotype-specificIR

PreexistingimmunityagainstaparticularAdserotypesignificantlyreducesthevector uptake shortens the duration of transgene expression and increasetoxicity

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 21: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Serotype switching to overcome neutralizing humoral immune response

Helper-Dependent Adenovirus Vectors Elicit IntactInnate but Attenuated Adaptive Host ImmuneResponsesInVivoagrave Improved safety and longer transgene expressionThe ability of preexisting antibodies to recognize epitopes on the capsid resulting in the vector clearance still remains a concern

Heterologous or chimeric viral surface proteinsEj HAd5 vector carrying a chimeric human-bovine fibers had a minimal hepatotoxicity and weaker inflammatory response upon iv injection in mice and also partially evaded the preexisting humoral immunityC o m b i n a t i o n o f f i b e r a n d h e x o n pseudotyping

PEGylation Decreased activation of the innate immune response (shild)

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 22: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

22

Human Herpes virus type 1

514

Herpesviruses

Family HerpesviridaeSubfamilies and Selected Genera

Examples

Alphaherpesviruses Simplexvirus Human herpes simplex virus type 1 and 2 Varicellovirus Varicella-zoster virusBetaherpesviruses Cytomegalovirus Human cytomegalovirus Roseolovirus Human herpesvirus 6 and 7Gammaherpesviruses Lymphocryptovirus Epstein-Barr virus Rhadinovirus Human herpesvirus 8

the family Malacoherpesviridae comprises viruses of oysters The family Herpesviridae listed here includes the well-known human pathogens that belong to all three subfamilies While some herpesviruses have broad host ranges most are restrict-ed to infection of a single species and spread in the population by direct contact or aerosols The hallmark of herpesvirus infections is the establishment of a lifelong latent or quiescent infection that can reactivate to spread to other hosts and often may cause one or more rounds of disease Many herpesvirus infections are not apparent but if the hostrsquos immune defenses are compromised infections can be devastating Some her-pesviruses are pathogens of economically important animals The study of herpesviruses has provided fundamental infor-mation about the assembly of complex virions the regulation of gene expression and mechanisms of immune system mod-ulation and insight into the biology of terminally differentiat-ed cells such as neurons

A

B

Long region(126 kb)

Short region(26 kb)

Lipid envelope

Envelope proteins(gBndashgN)(~12 proteins)

Nucleocapsid

dsDNAgenome

Tegument(gt20 proteins)

TRL UL

OriL OriS

IRL IRS TRSUS

OriS

Figure 13 Structure and genome organization of alphaherpesviruses (A) Virion structure Cryo-elec-tron tomograph of a slice through a single herpes simplex virus type 1 particle (Adapted from E Grunwald et al Science 3021396ndash1398 with permission) (B) Genome organization The herpes simplex virus type 1 genome can ldquoisomerizerdquo or recombine via the large inverted repeat sequences (TRL and IRL or IRS and TRS) such that all pop-ulations consist of four equimolar isomers in which unique long and short sequences (UL and US) are inverted with respect to each other There are at least 84 open reading frames in this 152-kbp genome as well as three origins of replication (Ori)

The order Herpesvirales currently consists of 3 families 3 subfamilies 17 genera and 90 species The family Alloher-pesviridae comprises fish and amphibian herpesviruses and

ASM_POV4e_Vol1_APPENDIXindd 514ASM_POV4e_Vol1_APPENDIXindd 514 72215 1224 PM72215 1224 PM

bull Large completely sequenced easy to manipulate genome

bull Half of the viral genome is composed of non- essential genes which can be replaced with heterologous genes

bull Efficiently infects a wide variety of mitotic and post- mitotic cells

bull The ability to do not integrate in the host genome

Why HSV-1 can be engineered for gene transfer

bull Enveloped virus bull Spherical to pleomorphic bull 150-200 nm in diameter

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 23: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

23

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

596

Figure 1 Schematic representation of the HSV-1 virion its genomic organization and the basic design of the HSV-1 derived amplicon plasmid (A) The mature wild-type HSV-1 virion is composed of 4 sub-compartments envelope tegument capsid and the 150-kb linear double-stranded DNA genome The envelope contains glycoprotein molecules involved in the cellular binding and viral entry processes of HSV-1 infection (B) The linear double-stranded DNA genome of wild-type HSV-1 encodes approximately 80-85 viral genes which are located in unique long (UL) and unique short (US) regions within the genome Inverted repeat elements (ab brsquoarsquo arsquocrsquo and ca) flank the unique regions and contain packaging signals required for cleavage and packaging of the replicated viral genome into virions and isomerization of the UL and US genomic segments Three viral origins of replication are located within the genome with 2 in the US region (oriS) and 1 in the UL region (oriL) Several viral genes whose functions are discussed are demarcated (C) The HSV-1 derived amplicon plasmid contains a single oriS or oriL and an ldquoardquo site and is devoid of all viral genes A bacterial origin of replication (ColE1) and an antibiotic resistant gene (Ampr) is included for bacterial propagation of the plasmid A transgene unit-of-interest can be cloned into the HSV-1 amplicon using standard molecular cloning techniques and packaged into HSV-1 amplicon viral particles using helper virus-based or helper virus-free packaging methodologies (Panel B was adapted from [17])

HSV-1 replicating vector recombinant vector with deletion of non-essential genes does not require specialized cell lines

Amplicon based HSV-1 vector is an amplicon-plasmid DNA containing the therapeutic gene The plasmid must contains an H S V - 1 o r i g i n o f r e p l i c a t i o n a n d t h e packaging signal (lsquoarsquo sequence)

H S V- 1 r e p l i c a t i o n defective vector contains deletions of essential f u n c t i o n s a n d c a n r e p l i c a t e s o n l y i n complementing cell lines

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 24: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

24

HSV-1-derived amplicon vectors bull Alberto Luis Epstein400

for the transduced organisms Furthermore the absence of virus genes in the amplicon genome strongly reduces the risk of reactivation complementation or recombina-tion with latent or resident HSV-1 genomes

Amplicons are quite versatile tools due to the fact that during their production the amplicon genome will replicate like HSV-1 via a mono-directional rolling circle-like mechanism generating long concatemers composed of tandem repeats of the amplicon plasmid (Boehmer amp Lehman 1997) (Fig 2B) Since infectious HSV-1 particles will always package approximately 150 Kbp DNA (the size of the virus genome) the number of repeats that a particular amplicon vector will carry and deliver will depend on the size of the original amplicon plasmid (Kwong amp Frenkel 1984) Therefore an ampli-con plasmid of around 5 Kbp will be repeated approx-imately 30 times in the amplicon vector while a very large amplicon plasmid carrying a 150 Kbp genomic locus will originate amplicon vectors carrying a single repeat of this sequence Actually a second major benefit that arises from the lack of virus genes in the amplicon plasmid is that most of the 150 Kbp capacity of the HSV-1 particle can be used to accommodate very large pieces of foreign DNA This is undoubtedly the most outstanding property of amplicons as there is no other available viral vector system displaying the capacity to deliver such a large amount of foreign DNA to the nuclear environment of mammalian cells apart from amplicons derived from other herpesviruses such as Epstein-Barr virus (EBV) or human cytomegalovirus (HCMV)

In this review we will describe the current state of the art of amplicon vector technology discuss the present limitations of these vectors and illustrate possible ways currently being investigated to improve the potential of these very interesting gene transfer tools

Production of amplicon vectors

Production of amplicon vectors involves the coordi-nated action of more than 50 viral proteins required to allow replication and packaging of the amplicon plasmid into fully infectious virus particles The viral genes en-coding these proteins could be supplied in principle by a helper virus by a viral genome or by a packaging cell line Although it is theoretically possible to construct a packaging cell line able to express the required pro-teins using a strict inducible system no such cells are yet available Hence the trans-complementing systems currently in use are based either on cloned viral DNA or on helper HSV-1 A major limitation long associated with the use of amplicons is the difficulty in producing large high-titre stocks of vector particles free of helper virus contamination and the improvement of methods to produce such stocks has been the concern of many studies over the past decade

The two major methods currently used for producing amplicons one based on infection with a HSV-1 helper virus and the other based on transfection of HSV-1 genes are described in Figs 3 and 4 respectively According to the first method which has long been the only method available cells were transfected with the amplicon plas-

Fig 1 the three types of HSV-1-based vectors Recombinant vec-tors are HSV-1 particles carrying an engineered HSV-1 genome Recombinant vectors can be attenuated (1) in which case they carry a mutation in a gene affecting virulence (white circle) and generally a reporter gene (in this case luciferase yellow arrow) to facilitate following the spread of the virus or defective (2) in which case they carry a deletion in at least one gene encoding an essential function (grey circle) Defective vectors generally carry a transgene of interest (red arrow) and a reporter gene (in this case GFP green arrow) to identify the infected cells Am-plicon vectors (3) are HSV-1 particles carrying a head-to-tail concatemer of a DNA derived from the amplicon plasmid in-stead of the virus genome These plasmids also generally carry a reporter gene and a transgene of interest In addition they carry one HSV-1 origin of DNA replication (oriS) and one packaging signal (pac) to allow amplification and packaging of the plasmid into HSV-1 particles

Fig 2 amplicon plasmid and amplicon vectors A an amplicon plas-mid is a standard Escherichia coli plasmid carrying one Herpes sim-plex virus type 1 (HSV-1) origin of DNA replication (oriS) one HSV-1 packaging signal (pac) and the transgenic and reporter sequences of interest (here represented as coloured arrows) B an amplicon vector is an HSV-1 particle carrying around 150 kbp of a head-to-tail con-catemer of DNA derived from the amplicon plasmid

Viruses 2009 1

605

Figure 3 Schematic representation of the latest packaging systems utilized for amplicon vector

production (A) The HSV-1LaLumlJ helper virus-based amplicon packaging technology involves

initial transfection of the transgene-harbored amplicon plasmid into an ICP4-complementing cell

line with subsequent superinfection with the HSV-1LaLumlJ helper virus which contains a unique

lsquofloxedrsquo packaging signal designed to reduce helper virus contamination The resultant viral

stock from the ICP4-complementing cell line that contains both packaged amplicon particles and

helper virus particles (at approximately a 11 ratio) is used to infect a second cell line which

expresses ICP4 and Cre recombinase Recombination of the loxP-sites via Cre recombinase

results in the deletion of the packaging signal from the helper virus which renders its own

genome packaging-defective and thereby reduces the titers of helper virus present (05) in the

final harvested amplicon vector stock (B) The helper virus-free packaging technology involves

the co-transfection of a permissive cell line such as Vero 2-2 cells with the transgene-harbored

amplicon plasmid and a bacterial artificial chromosome (BAC) designed to harbor the HSV-1

genome deleted in the packaging signals An accessory plasmid expressing other HSV-1

proteins such as vhs can be included to enhance titers The HSV-1-BACumlpac genome has been

strategically designed to preclude its packaging into viral particles thereby eliminating the

presence of helper virus contamination in the final amplicon vector stock (Figure was adapted

from [4568])

HSV-1 Amplicon system

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 25: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

25

bull HSV Amplicons

Advantages

bull Packaging potential is genome length

bull Multiple copies of transgene

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

Disadvantages

bull Contamination with helper virus and recombinants

bull Silencing

bull Lack scalable high titer vector manufacture

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 26: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

26

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 27: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

27

Essential genes are required to produce new infectious viralparticlesinpermissivecellcultureinfections

Accessory genes encode products that are not absolutelyrequiredincellculturebutareimportantforoptimumlyticreplicationoraffectthenaturallifecycleofthevirusinvivocontributingtohostrangepathogenesisorlatency

ICP27 were known to be essential for virus replication andthus deletion of either of these genes completely interruptedvirus growth at an early stage and provided the platform forthe first- and second-generation vectors (Fig 4B) Becausethese genes are essential a method for growing such viruseswas critical to vector engineering and production By themid-1990s cell lines capable of complementing both ICP4and ICP27 had been produced thus allowing production of avector backbone that only expressed the remaining IE genesICP0 ICP22 and ICP47 (Shepard and DeLuca 1991Marconi et al 1996) The deletion of both essential IEgenes prevented the occurrence of replication-competentrecombinants during complementation an important vectorsafety feature Fortunately we found that the 427 deletionvector was able to infect sensory nerves after intradermaldelivery (Goins et al 1994) The vector was transported tothe nerve cell body and the mutant virus genome becameestablished long-term but without the ability to reactivateMoreover it was possible to achieve transient gene ex-pression in neurons using strong promoters such as thehuman cytomegalovirus (HCMV) IE gene promoter Theremaining viral IE genes were rapidly silenced and theneurons appeared to be unharmed Although the HCMV

promoter was only transiently active in the ganglia the virallatency gene promoter remained active as evidenced by theexpression of LAT suggesting that these vectors achieved alatent-like state These HSV mutants represented the firstvectors useful for gene transfer to peripheral nerves openingup the door for gene therapy

A second breakthrough came from Bill Goins in ourgroup who discovered a sequence in the latency gene locusdownstream of the latency promoter that possessed inde-pendent promoter activity Bill referred to this element asthe second latency active promoter (LAP2) LAP2 wasmoveable and capable of expressing transgenes at genomiclocations outside of the latency locus and for extended times(Goins et al 1994) The discovery of LAP2 and the capa-bility of constructing ICP427 deletion vectors togetherprovided a vector system useful for long-term gene therapyof the brain and sensory nerve conditions (Glorioso et al2003)

Despite these advances our vectors were still cytotoxic tocells in culture and failed to persist in nonneuronal tissuesthereby limiting the broad utility of HSV vectors ICP0 inparticular caused cell cycle arrest and apoptosis (Samaniegoet al 1997) While removal of the remaining HSV IE genes

FIG 4 Cascade expression of HSV genes (A) The viral tegument protein VP16 (purple) activates the IE gene promoters(green) to express the IE gene products (blue) that in turn activate the promoters of the E genes (yellow) The E geneproducts replicate the viral DNA releasing expression of the L gene products (red) that package viral genomes de novo andform the virus particle structure (B) ICP4 (diploid) and ICP27 are essential IE genes Expression of E and L genes isblocked in replication-defective HSV deleted for ICP4 andor ICP27 Vector production requires the use of engineeredcomplementing cells First-generation replication-defective HSV vectors are deleted for ICP4 (orange) Second-generationvectors are deleted for both ICP4 (orange) and ICP27 (white) Third-generation vectors are functionally deleted for all ofthe IE genes ICP4 ICP27 ICP0 (diploid) (green) ICP22 (green) and ICP47 (not shown) and are noncytotoxic or interferewith cell division (C) Genomic structure of the third-generation vector deletion of the joint sequence and all remaining IEgenes blocks the expression of any viral genes requiring complementation of ICP4 ICP27 and ICP0 in engineered U2OScells

86 GLORIOSO

1erageneracioacuten(ICP4)

2dageneracioacuten(ICP4yICP27)

3erageneracioacuten

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

HSV-1 replication defective vector

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 28: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

28

HSV-1 replication defective vector gtcan replicates only in complementing cell lines

gtcontains deletions of essential functions

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

gtfail to ex- press any viral genes are completely devoid of toxicity and can persist in any nondividing cell type both in vitro and in vivo

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 29: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

29

HSV-1 replicating vector gt recombinant vector with deletion of non-essential genes gtremoval of accessory genes that contribute to neuropathogenesis gt late gene designated ICP345

gtdoes not require specialized cell lines gtconditionally replicating HSV vectors

1987 McGeoch et al 1988 Perry and McGeoch 1988 Wuet al 1988) we were nevertheless able to take advantage ofan important aspect of HSV biology cascade gene regula-tion (Fig 4A) Together the first wave of genes (immediateearly [IE] functions) provided activities to overcome innate

immune responses hide infected cells from immune sur-veillance block cell division prevent cell suicide preventrepression of viral gene expression and activate the ex-pression of early (E) and late (L) genes during progressionof the virus replication cycle Two of the IE genes ICP4 and

FIG 2 Genome structure loca-tion of gene classes (A) Diagram ofthe linear viral genome Blue boxesinverted repeats Terminal repeat(TR) and internal repeat (IR) flank-ing the long (L) and short (S) uniquesegments (B) The viral transcriptionunits are color coded as immediateearly (IE green) early (E yellow)early-late (L1 red) late (L2brown) and latency (LAT) gene(purple) The blue boxes representthe repeat regions that can be ma-nipulated to construct replication-defective vectors The internal jointregion contains genes that are du-plicated in the terminal repeats andcan be deleted for insertion of for-eign sequences The IE genes aredeleted to create completely silentvectors The LAT region containselements that can be used for inser-tion of transgenes that remain activein the absence of the IE genes

FIG 3 HSV vector design strategies (A) Amplicon designs Amplicons are genome-length plasmid-like vectors thatcontain a viral origin (ori) and packaging (pac) sequences that allow amplification of transfected amplicon DNA andincorporation into virus particles for amplicon delivery to cells Replication-defective helper virus supplies viral functionsneeded to replicate amplicon DNA and to make helper and amplicon particles in complementing (blue) cells Cotransfectionof noncomplementing cells (green) with amplicon DNA and helper DNA lacking packaging sequences will produce helper-free amplicon particles (B) Replication-defective vector designs Replication-defective viruses are deleted for one or moreessential virus genes and must be grown on cell lines that complement these deleted functions in trans (blue) Infection ofnoncomplementing cells in culture will not produce virus (green) and infection of other tissues (eg brain) will not allowvirus replication (C) Replication-competent (attenuated) vector designs These vectors can be propagated on non-complementing cells (green) and attenuated by removal of neuropathogenic accessory genes for targeting of brain tumorswithout harming normal brain tissue

HSV GENE VECTORS 85

186 TINS Vol 23 No 5 2000

attractive candidates for cell targeting because manytypes of neurones are defined by their neurotransmittersFor example the preproenkephalin and the tyrosine-hydroxylase (TH) promoters have been used to restrictexpression to enkephalinergic and catecholaminergicneurones respectively3637

Another approach to the problem might be to ma-nipulate the virus envelope to achieve cell-specific genedelivery HSV-1 viruses engineered to replace proteinsthat mediate attachment with the erythropoietin hor-mone peptide have been demonstrated to target erythro-poietin-receptor-bearing cells preferentially38 Theo-retically this approach can be used to direct HSV vectorstowards neurones that are characterized by specificreceptor types (for example neuropeptide receptors)Expression of multiple genes

The construction of multigene vectors is also a com-plex task Strategies to express multiple transgenes fromthe same vector are either founded on the use of mul-tiple promoters (that is separate expression of eachgene)3940 or on the insertion of IRES between the trans-genes (that is transcription of multiple genes from asingle promoter)4142 Model multigene vectors havebeen constructed with four or five independent trans-genes inserted at distinct loci which produces simul-taneous expression of all transgenes for up to sevendays40 Taking advantage of all available sites it is theo-retically possible to construct replication-defective vec-tors that express as much as 50 kb of foreign DNA Thesedata demonstrate the potential to create differenttransgene combinations for studies of their biologicalinteractions within nerve cells

Molecular analysis of behavior using HSV-1 vectors

Viral vectors have already been employed to study theeffect of alterations in the expression of single genes

within small groups of neurones on animal behaviourand disease models In this section the results obtainedusing HSV vectors will be focused on but examples ofthose obtained with other viral vectors will also bedescribedDrug addiction

Chronic drug administration elicits molecular adap-tations at the level of specific neuronal populations4344For example repeated exposure to morphine sensitizesanimals to its stimulant and rewarding properties45 andincreases expression of the gene for the AMPA gluta-mate-receptor subunit GluR1 in the ventral tegmentalarea (VTA) a crucial participant in reward mechanisms46The use of viral vectors has made it possible to investi-gate whether some of these adaptations correlate withspecific behavioural manifestations of addiction Thegene for GluR1 has been transfected into VTA neuronesusing an HSV-1 amplicon47 Thereafter sensitization tothe stimulant and rewarding properties of morphine hasbeen evaluated using appropriate protocols (morphine-induced locomotor activity and conditioned placepreference) Sensitization to both the stimulant and tothe rewarding properties of morphine was increasedafter vector-mediated expression of the gene for GluR1(but not after expression of other related genes) in theVTA This amplicon was not significantly neurotoxicand its behavioural effects were transient with a time-course that paralleled that of transgene expressionThus the behavioural consequences of morphine pre-exposure are potentiated by HSV-1-vector-mediatedexpression of the gene for GluR1 in the VTA

The repeated administration of cocaine also causescomplex molecular adaptations in the reward systemsFor example it increases phosphorylation of the tran-scription factor CREB (cAMP-response-element-bindingprotein) in the nucleus accumbens shell one of the

M Simonato et al ndash Herpes simplex vectorsT E C H N I Q U E S

trends in Neurosciences

(a)

(b)

ICP0 ICP0

ICP27 ICP4 ICP4

LATICP22

LAT

ICP3

45

UL2

UL3

UL4

UL1

UL5

UL6

UL7

UL8

UL9

UL2

2

UL2

4U

L25

UL2

7U

L28

UL2

9

UL2

2

UL2

5U

L26

UL2

7U

L28

UL2

9U

L30

UL3

1U

L32

UL3

3U

L34

UL3

5U

L36

UL3

7U

L38

UL4

UL4

8U

L49

UL5

3U

L52

US6

UL1

0U

L11

UL1

3

UL1

6

UL2

0U

L21

UL2

3U

L24

UL3

9U

L40

UL4

1

UL4

3U

L44

UL4

5U

L46

UL4

7

UL5

0U

L51

UL5

5U

L56

ICP3

45

US2

US3

US4

US6

US7

US8

US9

US1

0U

S11

Accessory

Essential

UL USIRL IRS TRSTRL

ICP22

ICP4

ICP0

ICP27

IE

VP16

E L+

+++

+ +

+

ndash

Fig 2 HSV-1 genome organisation and gene regulation (a) The virus genome The herpes simplex virus (HSV) genome is a linear double-stranded DNA molecule 152 kbin length and encoding at least 84 genes65 It is composed of two unique sequences the unique long (UL) and unique short (US) segments each of which is flanked by invertedrepeat elements (TRL and IRL IRS and TRS) The location of the essential genes which are required for viral replication in vitro and of the non-essential (accessory) geneswhich can be deleted without affecting replication in vitro are indicated Immediatendashearly (IE) genes (those that code for ICP4 ICP27 ICP22 and ICP0) and other locitargeted for genetic manipulation are highlighted (b) Temporal cascade of HSV gene expression detailing the role of HSV-1 IE gene transactivators and of the IE promoterstimulatory molecule VP16 The IE genes are expressed immediately after infection in the absence of de novo protein synthesis The VP16 virus tegument protein interactswith the cellular factor Oct1 to regulate the expression of the IE genes positively by binding to their promoters The IE-gene products ICP4 ICP27 and ICP0 are responsiblefor activating early (E) genes After viral DNA replication the ICP4 ICP22 and ICP27 IE polypeptides regulate expression of the late (L) genes ICP4 can also inhibit theexpression of IE genes including its own expression once transcription of the E class has been initiated

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 30: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Engineering techniquesSimple and highly efficient BAC recombineering usinggalK selectionSoslashren Warming Nina Costantino1 Donald L Court1 Nancy A Jenkins andNeal G Copeland

Mouse Cancer Genetics Program and 1Gene Regulation and Chromosome Biology LaboratoryNational Cancer Institute Frederick MD 21702-1201 USA

Received January 11 2005 Revised and Accepted February 4 2005

ABSTRACT

Recombineering allows DNA cloned in Escherichiacoli to be modified via lambda (l) Red-mediatedhomologous recombination obviating the need forrestriction enzymes and DNA ligases to modify DNAHere we describe the construction of three newrecombineering strains (SW102 SW105 and SW106)that allow bacterial artificial chromosomes (BACs) tobe modified using galK positivenegative selectionThis two-step selection procedure allows DNA to bemodifiedwithout introducing an unwanted selectablemarker at the modification site All three strains con-tain an otherwise complete galactose operon exceptfor a precise deletion of the galK gene and a defect-ive temperature-sensitive l prophage that makesrecombineering possible SW105 and SW106 cellsin addition carry L-arabinose-inducible Cre or Flpgenes respectively The galK function can be selec-ted both for and against This feature greatly reducesthe background seen in other negative-selectionschemes and galK selection is considerably moreefficient than other related selection methodspublished We also show how galK selection can beused to rapidly introduce point mutations deletionsand loxP sites into BAC DNA and thus facilitatefunctional studies of SNP andor disease-causingpointmutations the identificationof long-range regu-latory elements and the construction of conditionaltargeting vectors

INTRODUCTION

Bacterial artificial chromosomes (BACs) have become theDNA of choice for genomic sequencing due to their high

stability and large insert size (100ndash300 kb) (1) BACs arealso being used more and more for making transgenic micesince in many cases all of the important regulatory sequencesrequired for normal gene expression can be found on a singleBAC (23) Many laboratories also use BACs as the startingpoint for making gene-targeting constructs for manipulatingmouse genes using ES cell technology (knock-outs knock-insand conditional targeting using CreloxP) (45)

Recombineering (recombination-mediated genetic engi-neering) makes it possible to modify BAC DNA via homo-logous recombination [reviewed in (67)] Recombineering ismade possible through the use of three l Red-encoded genesexo bet and gam exo encodes a 50ndash30 exonuclease that pro-duces 30 overhangs from introduced double-stranded DNAtargeting cassettes (dsDNA) bet encodes a pairing proteinthat binds to the 30 overhangs and mediates its annealingand homologous recombination with complementary DNApresent on the BAC At the same time gam encodes an inhib-itor of the Escherichia coli RecBCD exonuclease and therebyprotects the linear DNA-targeting cassette from degradationby RecBCD l Red (or the corresponding RecE and RecTgenes of the prophage Rac) can be expressed from a multicopyplasmid using an inducible promoter (89) Alternatively thesegenes can be expressed from a stably integrated defectivel prophage where exo bet and gam are controlled by thestrong phage promoter pL under stringent control of thetemperature-sensitive repressor cI857 (1011) In the pro-phage system exo bet and gam are not expressed when thebacteria are kept at 32C By shifting the bacteria to 42C foras little as 15 min the genes are rapidly induced to very highlevels and homologous recombination is very efficient

Using recombineering one can easily subclone a genomicfragment from a BAC by gap repair either for use as atransgene directly or for subsequent manipulation to make agene-targeting construct The introduction of selectable mark-ers into a BAC is also very easy using recombineering How-ever a major limitation to the usefulness of BACs is the easeand efficiency with which one can make subtle and lsquoseamlessrsquo

To whom correspondence should be addressed at Mouse Cancer Genetics Program Center for Cancer Research National Cancer Institute West 7th Street at FortDetrick Bldg 539 PO Box B Frederick MD 21702-1201 USA Tel +1 301 846 1260 Fax +1 301 846 6666 Email Copelandncifcrfgov

ordf The Author 2005 Published by Oxford University Press All rights reserved

The online version of this article has been published under an open access model Users are entitled to use reproduce disseminate or display the open accessversion of this article for non-commercial purposes provided that the original authorship is properly and fully attributed the Journal and Oxford University Pressare attributed as the original place of publication with the correct citation details given if an article is subsequently reproduced or disseminated not in its entirety butonly in part or as a derivative work this must be clearly indicated For commercial re-use please contact journalspermissionsoupjournalsorg

Nucleic Acids Research 2005 Vol 33 No 4 e36doi101093nargni035

SW102 bacteria E Coli derived strain designed for BAC recombineering using galK positivenegative selection These bacteria contain a fully functional gal operon except the galK gene is missing The ability to grow on galactose minimal medium can be restored by adding galK in trans by inserting a galK expression cassette into a BAC Next the galK cassette can be replaced by any dsDNA carrying any desired mutation by selection against galK using 2-deoxy- galactose

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

galK expression cassette

Having produced a bacterial strain SW102 which is galKdefective the next step was to make a galK expression cassettethat could be used to restore the bacteriarsquos ability to grow ongalactose by providing galK in trans This was achieved byPCR amplification of the wild-type galK ORF from W3110cells We then added a minimal bacterial promoter em7 usinga two-step PCR approach (see Materials and Methods) andcloned the expression cassette into pBluescript SK We callthis plasmid pgalK (Table 1) The constitutively active galKexpression cassette can easily be amplified by PCRwith homology arms added to the primers (see Materialsand Methods)

Making a single base pair substitution

The general scheme for making mutations in BACs using galKselection is depicted in Figure 2 To test the galK selectionsystem for BAC recombineering we decided to introduce apoint mutation into a BAC containing the murine Nras gene(CITB clone 50J2) The sequence of the glycine-coding codon12 is GGT By changing this codon to GAT we would obtainthe desired mutation G12D In order to introduce this muta-tion into the BAC we first amplified the galK expressioncassette by PCR using primers with 50 bp of homology toeither side of the second position of the GGT codon Followinghomologous recombination this targeting would introduce a1 bp deletion into codon 12 in addition to inserting the galKselection cassette Instead of deleting the basepair the galKcassette could have been inserted right next to the basepairinstead SW102 cells containing the 50J2 BAC were heat-induced and made electrocompetent and then electroporatedwith the galK cassette Gal+ recombinant colonies were selec-ted for growth on galactose minimal medium with chloram-phenicol to maintain the BAC Bacteria grow more slowly onminimal media than on rich media and we generally pickcolonies after 2ndash3 days For this first step we do not expectany background colonies on the non-induced control plates ifthe pgalK plasmid is properly eliminated (see Materials andMethods) To purify the Gal+ colonies we streaked a fewcolonies on MacConkey galactose indicator plates to obtainsingle bright pinkred Gal+ colonies One of these single-cloned colonies was picked to initiate a culture for thenext step counterselection We find that there is no need toanalyze the Gal+ colonies further before proceeding tocounterselection

A 100 bp dsDNA oligo was then prepared by annealing twocomplementary oligos having 49 and 50 bp homologyrespectively to either side of the desired mutation a singleAT bp An aliquot of 200 ng of this oligo was then electro-porated into heat-induced and electrocompetent SW102 Gal+

cells containing the galK modified 50J2 BAC After electro-poration the bacteria were allowed 45 h outgrowth in a 32Cshaking waterbath The bacteria were then washed in M9 saltsto remove any rich medium and plated on minimal mediumwith glycerol DOG and chloramphenicol The 45 h out-growth is necessary to obtain complete segregation of therecombinant BACs containing the mutation After 3 dayswe obtained colonies with a ratio of 10ndash1001 when comparingplates with heat-induced to non-induced bacteria We picked12 colonies from the heat-induced plates for BAC minipreps

followed by SpeI restriction analysis (Figure 3A) Ten out ofthe twelve clones had the same restriction pattern as theunmodified 50J2 BAC DNA suggesting that the desiredreplacement of the galK gene by the point mutation hadoccurred This was confirmed by PCR amplification followed

Figure 2 Overview of the galK selection scheme The result of the firsttargeting event is the insertion of constitutively active galK into a definedposition on the BAC by selection on minimal medium containing galactoseand chloramphenicol to select for themaintenance of the BAC The bacteria arenow phenotypically Gal+ Next the galK cassette is replaced by a dsDNAoligoa PCR product or a cloned dsDNA fragment carrying a desired mutation(indicated by a star) and flanked by the same homology arms used in thefirst selection step This is achieved by negative selection using minimalmedium containing 2-deoxy-galactose (DOG) with glycerol as the sole carbonsource The bacteria become phenotypically Gal H1 and H2 homology arms1 and 2 respectively cat chloramphenicol acetyl transferase gene ori2 BACorigin of replication galK Ecoli galactokinase gene driven by a minimalpromoter

e36 Nucleic Acids Research 2005 Vol 33 No 4 PAGE 6 OF 12

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 31: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

31

Oncolytic Vectors

Advantages

bull Natural lytic cycle toxicity

bull Potential for enhanced virus spread within the tumor

bull Infection can be aborted with anti-HSV drugs

bull Can insert foreign genes

bull Ease of manufacture to high titer

bull Clinical experience is promising

Remaining Issues

Are current oncolytic viruses too attenuated

How best to achieve tumor targeting and improve initial vector distribution and methods of delivery

How to overcome the anti-viral response

How to induce anti-tumor immunity

How to combine with drug and radiotherapy

bull HSV Replication Defective

Advantages

bull Packaging potential is 40-50 Kb

bull Large or multiple transgenes

bull Removal of virus genes avoiding leakiness

bull Versatile construction in bacteria

bull High safety profile

bull Targeted Delivery and expression

Disadvantages

bull Transient gene expression

bull Highly defective viruses are difficult to manufacture

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 32: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

32

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 33: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Indica7onsampaddressedampbyampampgeneamptherapyampclinicalamptrials

httpwwwwileycomlegacywileychigenmedclinical

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 34: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Viral Vectors_ Vaccines

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 35: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

35

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29 Ledgerwood et al Chimpanzee Adenovirus Vector Ebloa Vaccine ndash Preliminary Report NEJM 26 November 2014 Rampling et al A Monovalent Chimpanzee Adenovirus Ebola Vaccine ndash Prelimiary Report NEJM 28January2015

NIHGSK Vaccine Candidate

bull Recombinant replication-defective Chimpanzee adenovirus type 3 (ChAd3)-derived vector encoding Ebolavirus Zaire (EBOV) glycoprotein (GP)

bull ChAds ndash evaluated as vaccine vectors in nonhuman primates and

human clinical trials ndash low world-wide seroprevalance in humans ndash not cross-neutralized by human anti-adenovirus sera

bull Immunological goal AEliginduction of effective antibody and CD8 T cell responses

bull Preclinical data AElig 100 protection in NHP model 5 weeks after a single dose of ChAd3-EBO-Z

bull Preliminary clinical data suggest vaccine has acceptable safety profile and is immunogenic

2

Chimpanzee Adenovirus expressing Ebolavirus Zaire Glycoprotein (ChAd3-EBO-Z vaccine)

Image credit Visualscienceru

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 36: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

36

NIHGSK Vaccine Candidate

4

Encouraging survival data in the Cynomolgus Macaque model

Acute protection ndash 1 dose Challenge at 5 weeks post vaccination

Stanley et al Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge Nature Medicine 201420(10)1126-29

EBOV IM challenge of 1000 PFU Mortality at 6-9 days

bull Acute protection associated with antibody responses

Ebolavirus challenge

0 1 2 3 4 5 Weeks

Vaccinate ChAd Ebola GP (n = 4group)

ChAd3-EBO-Z Vaccine Development

1 Scientific approachrationale - Vaccine candidate based on non replicative Chimpanzee adenovirus expressing GP of Ebola Zaire

(ChAd3-EBO-Z)

2 Supporting evidences - Protection data in NHP model 100 protection 5 weeks after single injection of ChAd3-EBO-Z - Acceptable safety and reactogenicity profile in Phase I studies - Highest dose induced statistically superior humoral immunogenicity after 1 dose with no difference

between populations

3 Current Status - NIH-sponsored efficacy study ongoing in Liberia

- Phase II part completed enrollment at 1500 (500 receiving ChAd3-EBO-Z) - Discussions ongoing about extending trial to Guinea

- Phase II safetyimmunogenicity study in West African Adults planned to start in June - Discussing with FDA and EMA approaches to confirm effectiveness if clinical efficacy is not feasible

11

Summary and Conclusion

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 37: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

37

EbolavirusampGPampvaccineampinampAdMVA

doi101038nm3702

IMchallenge GSKNIAIDNIH

Animalsinsingle-shotgroupswerevaccinatedwithChAd3atthedosesindicatedandexposedtoalethaldoseofEBOV10monthsaftertheprimevaccinationAnimalsinprime-boostgroupswereprimedwithChAd3boosted8weekslaterandexposedtoalethaldoseofEBOVasinthesingle-shotgroups

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 38: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

38

ChAd3-EBO-Z Dose decision 1x1011 vp

bull Analysis shared with Phase I investigators FDA EMA and African NRAsERCs

bull ChAd3-EBO-Z was shown to have an acceptable safety profile in Phase I ndash All doses are considered acceptable from an individual patient safety perspective ndash No suspected unexpected serious adverse reactions (SUSARS) reported ndash Majority of solicited adverse events that were reported were classified as mild ndash Local pain at the injection site headache and ldquoflu-likerdquo symptoms were the most common solicited adverse

events

bull ChAd3-EBO-Z was immunogenic in humans and demonstrated antigen-specific antibody responses against Ebola GP and T-cell responses

bull 1x1011 vaccine dose induced higher titers compared to the different lower doses based on both the observed or predicted values

8 Analysis contains unpublished data

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 39: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Ebola Vaccine

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 40: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

40

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 41: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Viral Vectors_ Viral Oncotherapy

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 42: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

42

Viruses 2016 8 9 3 of 22

Viruses2016ȱ8ȱ0000ȱ 3ȱofȱ22ȱ

ȱFigureȱ1ȱMechanismsȱofȱtumourȱdestructionȱthroughȱoncolyticȱvirotherapyȱOncolyticȱvirusesȱ(OVs)ȱmayȱexertȱ theirȱantiȬtumourȱeffectsȱ throughȱseveralȱmechanismsȱOVsȱ induceȱ theȱdeathȱofȱatȱ leastȱsomeȱ tumourȱ cellsȱ inȱ aȱdirectȱwayȱbyȱ infectingȱ theseȱ cellsȱandȱ replicatingȱ thereinȱProgenyȱvirusȱparticlesȱareȱreleasedȱandȱ infectȱneighbouringȱ tumourȱcellsȱresultingȱ inȱamplificationȱofȱ theȱ inputȱOVȱdoseȱ(boxȱ1)ȱOVsȱoftenȱinduceȱanȱimmunogenicȱcellȱdeathȱ(boxȱ2)ȱTumourȬassociatedȱantigensȱpathogenȬassociatedȱ molecularȱ patternȱ (PAMPs)ȱ andȱ dangerȬassociatedȱ molecularȱ patternsȱ(DAMPs)ȱareȱreleasedȱfromȱdyingȱtumourȱcellsȱandȱcomeȱintoȱcontactȱwithȱantigenȬpresentingȱcellsȱsuchȱasȱdendriticȱcellsȱ inȱ theȱ tumourȱmicroenvironmentȱLocalȱ inflammationȱasȱ inducedȱbyȱvirusȱinfectionȱstimulatesȱtheȱmaturationȱofȱdendriticȱcellsȱandȱtheirȱmigrationȱtoȱdrainingȱlymphȱnodesȱwhereȱ theyȱcanȱpresentȱ tumourȬassociatedȱantigensȱ toȱTȱcellsȱUnderȱoptimalȱconditionsȱ thisȱmayȱelicitȱanȱantiȬcancerȱCD4+ȱandȱCD8+ȱeffectorȱTȱcellȱresponseȱthatȱhasȱtheȱpotentialȱtoȱkillȱinfectedȱandȱuninfectedȱ tumourȱ cellsȱ Inȱ additionȱ someȱOVsȱ disruptȱ theȱ tumourȬassociatedȱ vasculatureȱ (viaȱinfectionȱofȱtumourȱendothelialȱcellsȱexpressionȱofȱantiȬangiogenicȱviralȱproteinsȱandorȱOVȬinducedȱinflammatoryȱresponses)ȱleadingȱtoȱischemiaȱandȱnecroticȱdeathȱofȱuninfectedȱtumourȱcellsȱ(boxȱ3)ȱ

VirusȬinducedȱ cellȱ deathȱ followingȱ infectionȱ andȱ inȬcellȱ multiplicationȱ isȱ aȱ complexȱmultifacetedȱprocessȱwhichȱisȱtriggeredȱmainlyȱbyȱcytotoxicȱviralȱproteinsȱalthoughȱthisȱhasȱyetȱtoȱbeȱstudiedȱforȱsomeȱoncolyticȱvirusesȱinȱuseȱTheseȱproteinsȱhaveȱmultipleȱmodesȱofȱactionȱwhichȱoftenȱdifferȱ fromȱ thoseȱ activatedȱbyȱ conventionalȱ cancerȱ therapiesȱAsȱ aȱ resultȱ theȱ likelihoodȱofȱcancerȱcellsȱacquiringȱresistanceȱtoȱOVȱtreatmentȱasȱseenȱwithȱotherȱantiȬcancerȱmodalitiesȱhasȱnotȱbeenȱdocumentedȱ soȱ farȱ (inȱ contrastȱ toȱ theȱ resistanceȱdevelopedȱ toȱotherȱantiȬcancerȱmodalities)ȱHoweverȱitȱcannotȱbeȱruledȱoutȱthatȱsomeȱcancerȱcellsȱmayȱacquireȱresistanceȱtoȱOVsȱthroughȱtheȱlossȱofȱcellularȱpermissivenessȱfactorsȱegȱcellȱsurfaceȱreceptorsȱthatȱareȱessentialȱforȱvirusȱuptakeȱorȱ theȱalterationȱofȱsignallingȱpathwaysȱ requiredȱ forȱ theȱvirusȱ lifeȱcycleȱConverselyȱexamplesȱofȱOVsȱovercomingȱcancerȱcellsrsquoȱdrugȱresistanceȱhaveȱfrequentlyȱbeenȱdocumentedȱ[12]ȱFollowingȱcellȱlysisȱprogenyȱvirusȱparticlesȱareȱreleasedȱandȱcanȱinfectȱneighbouringȱtumourȱcellsȱInȱthisȱmannerȱ

Figure 1 Mechanisms of tumour destruction through oncolytic virotherapy Oncolytic viruses (OVs)may exert their anti-tumour effects through several mechanisms OVs induce the death of at least sometumour cells in a direct way by infecting these cells and replicating therein Progeny virus particles arereleased and infect neighbouring tumour cells resulting in amplification of the input OV dose (box 1)OVs often induce an immunogenic cell death (box 2) Tumour-associated antigens pathogen-associatedmolecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) are released fromdying tumour cells and come into contact with antigen-presenting cells such as dendritic cells inthe tumour microenvironment Local inflammation as induced by virus infection stimulates thematuration of dendritic cells and their migration to draining lymph nodes where they can presenttumour-associated antigens to T cells Under optimal conditions this may elicit an anti-cancer CD4+

and CD8+ effector T cell response that has the potential to kill infected and uninfected tumour cellsIn addition some OVs disrupt the tumour-associated vasculature (via infection of tumour endothelialcells expression of anti-angiogenic viral proteins andor OV-induced inflammatory responses) leadingto ischemia and necrotic death of uninfected tumour cells (box 3)

Virus-induced cell death following infection and in-cell multiplication is a complex multifacetedprocess which is triggered mainly by cytotoxic viral proteins although this has yet to be studied forsome oncolytic viruses in use These proteins have multiple modes of action which often differ fromthose activated by conventional cancer therapies As a result the likelihood of cancer cells acquiringresistance to OV treatment as seen with other anti-cancer modalities has not been documented so far(in contrast to the resistance developed to other anti-cancer modalities) However it cannot be ruledout that some cancer cells may acquire resistance to OVs through the loss of cellular permissivenessfactors eg cell surface receptors that are essential for virus uptake or the alteration of signalling

Mechanisms of tumour destruction through oncolytic virotherapy

Oncolytic viruses are a new class of therapeutic agents that promote anti-tumour responses through two distinct mechanisms of action

i) selective replication within neoplastic cells resulting in a direct lytic effect on tumour cells and

ii) induction of systemic innate and tumour-specific adaptive immune responses

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 43: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Nature Reviews | Drug Discovery

Oncolyticvirus

ROS

ROS

bull ER stressbull Genotoxic stress

Cancer cell

Viraloncolysis

CD8+ T cell

CD4+ T cell

NK cell

Antigen presenting cell

Cytokinereceptors

Cytokinereceptors

CD28

Cytotoxicity

Cytotoxicity

Activation

TCR

TCR

IL-2R

IL-2

MHC MHCTLR

MHC

CD40

CD40L

bull Type I IFNsbull DAMPsPAMPs

DAMPsPAMPs

bull Type I IFNsbull Cytokines

bull Viral proteinsbull Viral genome

bull Viral antigensbull TAAsneoantigens

bull CD80CD86bull Chemokine receptors

PAMPsbull Viral capsidsbull Viral DNAbull Viral dsRNAssRNAbull Viral proteins

DAMPsbull HSPsbull HMGB1bull Calreticulinbull ATPbull Uric acid

Cytokinesbull Type I interferonsbull TNF_bull IFNabull IL-12

Infection

Release Release

Antigenuptake

Releasesecrete

likely to be permissive to infection313637 Stromal cells such as cancer-associated fibroblasts may be infected by oncolytic viruses but are non-permissive to viral replication Thus fibroblasts may act as a decoy reservoir for oncolytic viruses reducing the delivery of infectious virions to cancer cells38 Another mechanism that may limit the overall effectiveness of oncolytic viruses is the susceptibility of cancer cells to apoptosis which may be induced by viral infection or other factors39 If cells undergo apoptosis too rapidly this will reduce the time

for viral replication and propagation and decrease the amount of active virus in the tumour ultimately limiting the active intratumoural dose

Development of oncolytic viruses as drugsAs oncolytic viruses are live viral particles the over-all design of oncolytic virus strategies must consider approaches to tumour cell targeting and attenuating viral pathogenesis as well as approaches to limit viral immunogenicity while promoting tumour cell killing

Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses The therapeutic efficacy of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti-tumour immune responses Upon infection with an oncolytic virus cancer cells initiate an antiviral response that consists of endoplasmic reticulum (ER) and genotoxic stress This response leads to the upregulation of reactive oxygen species (ROS) CPFVJGKPKVKCVKQPQHCPVKXKTCNE[VQMKPGRTQFWEVKQP415CPFE[VQMKPGUURGEKHKECNN[V[RG|+KPVGTHGTQPU+(0UCTGTGNGCUGF from the infected cancer cell and stimulate immune cells (antigen presenting cells CD8+6|EGNNUCPFPCVWTCNMKNNGT0-cells) Subsequently the oncolytic virus causes oncolysis which releases viral progeny pathogen-associated molecular patterns (PAMPs) danger-associated molecular pattern signals (DAMPs) and tumour associated antigens (TAAs) including neo-antigens The release of viral progeny propagates the infection with the oncolytic virus The PAMPs (consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering activating receptors such as Toll-like receptors (TLRs) In the context of the resulting immune-stimulatory environment TAAs and neo-antigens are released and taken up by antigen presenting cells Collectively these events result in the generation of immune responses against virally infected cancer cells as well as FG|PQXQ immune responses against TAAsneo-antigens displayed on un-infected cancer cells CD40L CD40 ligand dsRNA double-stranded RNA HMGB1 high mobility group box 1 HSP heat shock protein IL-2 interleukin-2 IL-2R IL-2 receptor MHC major histocompatibility complex ssRNA single-stranded RNA TCR T cell receptor TNFα tumour necrosis factor-α

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 647

copy 2015 Macmillan Publishers Limited All rights reserved

Mechanisms of oncolytic virus immunotherapy a Healthy cell

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSVbull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpes bull Adenovirusbull Vacciniabull+PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

IFNRIFN

IFN

IFNR

b Cancer cell

Cytoplasm

Nature Reviews | Drug Discovery

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

PKR

MYD88

NF-gB IRF7 IRF3

TRIFRIG-1

TRAF6 TRAF3

bull VSV bull NDVbull Measlesbull Vaccinia

bull IRF7bull PKR

bull Reovirusbull Herpesbull Adenovirus bull Vacciniabull +PȯWGPC

Translation

Apoptosis

NDV

NDV NDV

JAK

STAT

IRF9

STAT

IRF9

Type IIFNs

IRF7

IRF3

Pro-KPȯCOOCVQT[cytokines

NF-gB

dsRNA

Viral TLRligands

TLR

darr darr

darr

darr

darr

darr

Nucleus

Different viruses can also manipulate distinct aberrant signalling factors within tumour cells to block apoptosis which allows more time for the virus to complete its life cycle Following viral replication most oncolytic viruses induce cell death which can directly eliminate viable tumour cells but also sets the stage for initiating systemic immune responses Induction of host immune responses can be greatly aided by both the type of cell death and the

release of danger signals from virus-infected cells For example necrosis or pyroptosis are more immunogenic forms of cell death than apoptosis

Induction of systemic anti-tumour immunity The induction of systemic innate and tumour-specific adaptive immune responses appears to be a critical element for tumour eradication with oncolytic viruses Following oncolytic

Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways a | Following viral infection most normal cells activate an antiviral pathway that allows to contain viral infections The antiviral machinery can be triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1) Once a virus is detected a signalling ECUECFGVJTQWIJUGXGTCNV[RG|+KPVGTHGTQP+(0GNGOGPVUCPWUMKPCUG- signal transducer and activator of transcription (STAT) and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that limits viral spread and can target infected cells for apoptosis or necrosis Local IFN production induced by the innate immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR) TLRs signal via the myeloid differentiation primary response protein MYD88 TIR-domain-containing adapter-inducing IFNβ (TRIF) IRF7 IRF3 and nuclear factor-κB (NF-κ$KPFWEKPIVJGRTQFWEVKQPQHRTQKPHNCOOCVQT[E[VQMKPGUCPFV[RG|++(0U 6JGV[RG|++(0UUKIPCNVJTQWIJVJG-s566UKIPCNNKPIRCVJYC[TGUWNVKPIKPVJGWRTGIWNCVKQPQHEGNNE[ENGTGIWNCVQTUUWEJCURTQVGKPMKPCUG42-4CPF+4(YJKEJNKOKVXKTCNURTGCFD[DKPFKPIVQXKTCNRCTVKENGUCPFVTKIIGTKPIV[RG|++(0transcriptional pathways promoting abortive apoptosis of infected cells and the production of cytokines that alert the immune system to the presence of a viral infection b | In cancer cells however this process is disrupted Cancer cells may downregulate key signalling components within the innate signalling pathway including RIG-1 IRF7 and IRF3 (REF 1) This limits detection of viral particles by TLR and RIG-1 making cancer cells more susceptible to viral TGRNKECVKQP(WTVJGTOQTGECPEGTEGNNUOC[FQYPTGIWNCVGMG[EQORQPGPVUQHVJGV[RG|++(0UKIPCNNKPIRCVJYC[2ndash7 VJGTGD[NKOKVKPIVJGRTQCRQRVQVKECPFEGNNE[ENGTGIWNCVQT[GHHGEVUQHV[RG|++(0UNVJQWIJFCVCCTGNKOKVGFVJGHKIWTGdepicts individual viruses near the factors andor pathways that are known to promote viral elimination in normal cells (part a) or that support viral replication owing to factor deficiency in cancer cells (part b) dsRNA double-stranded RNA NDV Newcastle disease virus TRAF TNF-associated factor VSV vesicular stomatitis virus

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 645

copy 2015 Macmillan Publishers Limited All rights reserved

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 44: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

44

Development of oncolytic viruses as drugs

Targeting oncolytic viruses to cancer cells

Exploiting aberrant signalling pathways in cancer

Augmenting anti-tumour immunity

Enhancing lytic activity

Limiting antiviral immune responses

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 45: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Nature Reviews | Drug Discovery

HVEMCAR

ICAM-1

NectinIntegrins

LDLR

SARs

DAF

CD155

CD46

SLAM

Poliovirus

Measlesvirus NDV

VSV

Seneca ValleyVirus

Coxsackievirus

Adenovirus

Vaccinia virus

Herpesvirus

Parvovirus

Reovirus

prostate cancer63 This virus does not produce E1A in healthy cells and these cells undergo apoptosis thereby restricting viral proliferation in healthy tissue However in prostate cancer cells the PSA promoter is highly active and E1A is selectively expressed resulting in proliferation of the adenovirus and virus-mediated cell lysis In a similar manner the oncolytic adeno-virus KH901 was engineered to express E1A under the control of the human telomerase reverse transcriptase (TERT) promoter which is activated in a large num-ber of cancers64 Furthermore two binding sites of the transcription factor E2F1 (a regulator of the cell cycle) were included in the TERT promoter to restrict prolif-eration to actively dividing cells Another example is the oncolytic adenovirus CG0070 for which replication is restricted to retinoblastoma (Rb)-defective cells (a com-mon mutation in cancer) because E1A expression was placed under the regulation of the E2F1 promoter65 In healthy cells Rb inhibits E2F thus inhibiting E1A transcription66 Furthermore adenoviruses have been engineered to selectively replicate in hypoxic environ-ments such as those found inside tumours by placing the E1A gene under transcriptional regulation by the

hypoxia-induced transcription factor HIF-1α this strategy was shown to be effective in a murine xenograft model of glioma67 Several oncolytic adenoviruses includ-ing ONYX-015 and H101 have been designed with a deletion in the gene coding for the protein E1B which can bind to and inactivate the pro-apoptotic protein p53 (REFS 8586) Thus healthy cells that are infected with these viruses can undergo p53 - mediated abor-tive apoptosis whereas cancer cells that commonly inactivate p53 remain susceptible to viral infection Furthermore these adenoviruses preferentially prolifer-ate in and lyse p53-deficient cancer cells which account for nearly 50 of all cancers

Another approach to improve post-entry tumour specificity has been to encode synthetic miRNA target-ing sequences (miRTS) into the 3ʹ untranslated region (UTR) of the fusion gene of the measles virus These sequences bind cellular microRNAs (mi RNAs) and repress viral replication Because normal and cancer cells exhibit differential expression of cognate miRNA elements the engineered oncolytic virus with miRTS can be blocked from replicating in normal cells where specific mi RNAs are expressed Such a construct was

Figure 3 | Mechanisms of viral entry into cancer cells Oncolytic viruses utilize several mechanisms to enter host cells including cell surface receptors that are frequently overexpressed on cancer cells Some viruses are able to via more than one receptor and some receptors can promote the entry of more than one type of virus Some viruses use endocytosis through membrane fusion and syncytia formation to enter cells Certain oncolytic viruses are known to preferentially target cancer cells but the cell surface receptor for entry has not been identified CAR coxsackievirus-adenovirus receptor DAF decay accelerating factor HVEM1 herpesvirus entry mediator 1 ICAM-1 intercellular adhesion molecule 1 LDLR low-density lipoprotein receptor NDV Newcastle disease virus SARs sialic acid receptors SLAM signalling lymphocytic activation molecule VSV vesicular stomatitis virus (VSV)

REV IEWS

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | SEPTEMBER 2015 | 649

copy 2015 Macmillan Publishers Limited All rights reserved

overexpressedonsomecancercells

includingmelanomaand

variouscarcinomasoverexpressedincancerssuchas

multiplemyelomamelanomaandbreastcancer

ectopicallyexpressedincertaincancers

suchasglioblastomamulbformeandplaysanimportantroleintumorcellmigrabon

invasionandmetastasis

Targeting oncolytic viruses to cancer cells

Natural tropism for cell surface proteins that a r e a b e r r a n t l y expressed by cancer cells

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 46: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Tumoramptarge7ngbull ReceptortargeMng

AltermeaslesvirusHAtotargettumormarkers

HSVglycoproteinDengineeredtocontainIL13orsinglechainanMbodiesagainsthumanepithelialgrowthfactorreceptor2ongliomasandbreasttumors

AdenovirusinserMonofdomainsthatrecognizetumorAgintofiber

Hexoninterlacingprotein

Adaptorsthatbindfiberandretarget

to express a single-chain anti- body that recognizes carcinoembryonic antigen (CEA) a tumour antigen that is selectively expressed on certain adenocarcinomas

Ad3Fiber

Ad3

Ad3Fiber

Enhances the infectivityo n h uma n o v a r i a nadenocarcinoma cel llinesandprimaryhumanovariancancercells

OV can also be engineered to directly target unique cell surface receptors expressed by cancer cells

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 47: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

47

Post8entryamptarge7ng

doi101038nrmicro3140

selectively promotes the expression of viral genes (ej E1A) or engineered transgenes in target cells using tumour- specific transcriptional control

restricts infection in non-target cells using tissue-specific miRNAs that recognize target sequences that have been engineered into oncolytic virus genomes (UTR)

Tumour-associated

restrict virus replication in off-target tissues

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 48: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

E1b55k-deleted Ad (H101 or Onyx-015)

normal cell

Ad ΔE155k

tumor cell (p53 mutated or deleted) Ad ΔE155k

Apoptosis before viral replication

p53p53

p53

E155k

Apoptosis inhibitionp53

Only replicates in p53 deficient tumors

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 49: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

49

Armingampviralampvectors

bull EnhancetherapeuMcefficacyofoncolyMcvirushardtoinfect100ofcells

bull Strategiesthatkilltumorcellssurroundingthoseinfectedbystanderkilling

bull Prodrugconvertases

bull Iontransportprotein

bull ImmunosMmulatoryfactors

Arming viral vectors

lsquobystander cell killingrsquogt a protein that is expressed by the ov sensitizes both the infected cell and surrounding uninfected cells to subsequent combination therapies or immune destruction

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 50: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

50

Armingampwithampprodrugampconvertases

bull Thymidinekinaseconvertsganciclovirtoganciclovirtriphosphate

bull Cytosinedeaminaseconverts5fluorocytosineto5fluorouracil

bull ThesenucleosideanaloguesstopDNAreplicaMonoftumorcells

httpwwwncbinlmnihgovpubmed24292552

PNP purine nucleoside phosphorylase gt fludarabine phosphate into 2-fluoro- adenine

to activate non-toxic precursors which generate highly toxic metabolites in the tumour microenvironment

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 51: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

51

Arming with immnunoestimulatory factors

bull Expression of GM-CSF to stimulate the production of granulo- cytes and monocytes which in turn stimulate adaptative immunity against tumour-associated antigens

Nature Reviews | Microbiology

Prodrug activationa

Prodrug

Toxic metabolite

Bystander cells

No treatment Virotherapy

Radioactive iodide

Immune cell

Activated immune cell

Stimulatory peptides

NIS

Uninfected tumour cell Dying tumour cell

Virus

Radiosensitizationb

Immunostimulationc

Ionizing radiation

γ-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy electromagnetic γ rays

β-emitting isotopesRadioactive isotopes that emit ionizing radiation in the form of high-energy high-speed electrons or protons

out of the infected cell and into surrounding cells98103 to induce chemotherapeutic bystander killing (FIG 4a) making them clinically relevant arming strategies for virotherapy We focus in this section on the most clini-cally advanced viruses that are armed with the cytosine deaminase and PNP transgenes as well as on promis-ing preclinical viruses that are ready to enter future clinical trials

Cytosine deaminase and PNP have both been incorporated into several virus classes that have been preclinically tested including viruses that are based on Herpesviridae104 Adenoviridae105 Poxviridae106107

Paramyxoviridae108109 and Rhabdoviridae110 The most advanced cytosine deaminase virus is a replication-competent retrovirus known as Toca 511 which inte-grates the cytosine deaminase transgene into the genome of infected cells to establish permanent reservoirs of tumour cells that are sensitive to subsequent rounds of chemotherapy using 5-FC19 Toca 511 is currently being tested in combination with 5-FC in Phase I and II clini-cal trials using intratumoural administration in patients with grade 4 glioblastoma multiforme (TABLE 1) HSV and VSV viruses that express cytosine deaminase are also being preclinically developed for combination therapies using 5-FC104110

Intratumoural administration of an adenovirus with the PNP transgene in combination with intrave-nous fludarabine has been used to treat patients with head and neck tumours111 (clinicaltrialsgov identifier NCT01310179) In general cytosine deaminase and PNP transgenes can be applied to many virus fami-lies because their small sizes and low cellular toxicities incur minimal negative effects on in vitro virus fitness or production However the timing of prodrug dosing in vivo must be optimized to ensure that virus repli-cation and spread is sufficient for maximal synergistic effects with the chemotherapeutic prodrug104109112 In this respect a PNP-expressing measles virus that has been retargeted to CD20 has been extensively tested in combination with fludarabine in preclinical models of lymphoma108109

Radiosensitization The normal physiological func-tion of the human sodiumndashiodide symporter (NIS) is to transport iodide ions into cells which occurs pre-dominantly in the thyroid but also in the stomach salivary glands and mammary glands113114 When NIS is expressed from the genome of an oncolytic virus infected cells concentrate iodide or similar isotopes intracellularly During virotherapy γ-emitting isotopes such as 123I and pertechnetate can be administered to visualize virus replication using single-photon emis-sion computed tomography (SPECT BOX 2) whereas β-emitting isotopes such as 131I and 188Re can be admin-istered to specifically induce radiation poisoning within the tumour microenvironment (FIG 4b) in analogy to the clinically well-established radiotherapy that is used for metastatic thyroid cancer

The NIS transgene system has undergone extensive preclinical development in multiple virus families and radiovirotherapy has consistently achieved synergistic tumour destruction in several radiosensitive preclini-cal disease models115ndash117 Measles virus is an especially efficacious virus for NIS-mediated imaging (BOX 2) and radiovirotherapy Preclinical studies of lymphoma118 ovarian cancer119 myeloma120 and mesothelioma121 in addition to many other disease models117 have used NIS expression and SPECTndashcomputed tomography (SPECTndashCT) imaging to visualize and quantify virus replication and enhance disease regression using com-bination radio virotherapy Phase I clinical trials using NIS-expressing measles virus have been initiated for ovarian cancer myeloma mesothelioma and head and

Figure 4 | Arming strategies that induce bystander cell killing a | Convertase enzymes that are expressed in infected cells metabolize prodrugs into toxic metabolites that diffuse and kill uninfected tumour cells b | The sodiumndashiodide symporter (NIS) concentrates radioactive ions in infected cells which induces radiation poisoning of uninfected bystander tumour cells c | Immunostimulatory transgenes that are expressed in infected cells prime responses against tumour antigens which causes the systemic destruction of tumour cells

REVIEWS

30 | JANUARY 2014 | VOLUME 12 wwwnaturecomreviewsmicro

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 52: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

52

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 53: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

Figure1Schematicdiagramshowingtheherpessimplextype1virusfromwhichT-VECwasdeveloped(A)andthemodifiedvectorknownastalimogenelaherparepvec(T-VEC)(B)ViralICP345neurovirulencegeneandtheICP47immunogenicitygenehavebeendeletedinT-VECThehumanGM-CSFgenedrivenbyaCMVpromoterhasbeeninsertedintotwodeletedICP345lociandthedeletedICP47resultsinearlyactivationoftheUS11promoterAbbreviationsIRinternalrepeatLlongSshortTRterminalrepeatUuniqueGM-CSFgranulocyte-macrophagecolonystimulatingfactorT-VECtalimogenelaherparepvecCMVcytomegalovirusICPinfectedcellproteinpApolyAtailUSuniqueshort

BasicbiologyofT-VEC

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 54: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

54

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 55: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

ONCOS 02

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 56: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

56

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved

Page 57: Viral vectors - Instituto de Investigaciones ... · Viral vectors are used for the delivery of genetical material into cells.Because viruses have a natural ability to efficiently

57

of replicating cells resulting in the termination of DNA synthesis and ultimately cell death97 In this system the expression of TK is restricted to cells with an active osteo-calcin promoter and this increases their susceptibility to treatment with the thymidine analogue ganciclovir However as ganciclovir can block viral replication this approach may inhibit oncolytic virus activity9899

Two other suicide genes that have been tested are bacterial cytosine deaminase (CD) and adenovirus death protein (ADP)100101 CD can transform 5-fluoro-cytosine into 5-fluorouracil (5-FU) which is cytotoxic ADP is a nuclear membrane glycoprotein that is required in the late stages of adenovirus infection for efficient cell lysis and release of viral particles Enhanced

Table 3 | Key oncolytic viruses in clinical trials

Virus Manufacturer Modification Number of clinical trials Cancers

2JCUG|+ 2JCUG|++ 2JCUG|+++

Adenovirus

Onyx-015 Onyx Pharmaceuticals

6[RG|EJKOCGTC E1B deletion

6 6 0 Head and neck cancer pancreatic cancer ovarian cancer colorectal cancer gliomas lung metastases and liver metastases

H101 Shanghai Sunwaybio

E1B deletion partial E3 deletion

1 2 1 Squamous cell carcinoma and head and neck cancer

DNX-2401 DNAtrix Δ24-RGD insertion 4 0 0 Glioblastoma ovarian cancer

VCN-01 VCN Biosciences PH20 hyaluronidase insertion

2 0 0 Pancreatic cancer

Colo-Ad1 PsiOxus Therapeutics

Chimeric Ad113 group B 1 2 0 Colon cancer NSCLC renal cancer bladder cancer and ovarian cancer

ProstAtak Advantagene 6-KPUGTVKQP 4 1 1 Pancreatic cancer lung cancer breast cancer mesothelioma and prostate cancer

Oncos-102 Oncos Therapeutics

Δ24-RGD-GM-CSF insertion 1 0 0 Solid cancers

CG0070 Cold Genesys GM-CSF and E3 deletion 1 1 1 Bladder cancer

Vaccinia virus

Pexa-vac

GPPGTGZBiotherapeutics

)5(KPUGTVKQP6-disruption

7 6 0 Melanoma liver cancer colorectal cancer breast cancer and hepatocellular carcinoma

GL-ONC1 Genelux 6-FKUTWRVKQPJCGOCIINWVKPdisruption F145L disruption

4 1 0 Lung cancer head and neck cancer and mesothelioma

Herpesvirus

T-VEC Amgen ICP345 deletion US11 deletion GM-CSF insertion

2 3 2 Melanoma head and neck cancer and pancreatic cancer

G207 Medigene ICP345 deletion UL39 disruption

3 0 0 Glioblastoma

HF10 Takara Bio UL56 deletion selected for single partial copy of UL52

2 1 0 Breast cancer melanoma and pancreatic cancer

SEPREHVIR (HSV1716)

Virttu Biologics ICP345 deletion 5 1 0 Hepatocellular carcinoma glioblastoma mesothelioma neuroblastoma

OrienX010 OrienGene Biotechnology

ICP345 deletion ICP47 deletion GM-CSF insertion

1 0 0 Glioblastoma

Reovirus

Reolysin Oncolytics Biotech

None 15 9 0 Glioma sarcomas colorectal cancer NSCLC ovarian cancer melanoma pancreatic cancer multiple myeloma head and neck cancer

Seneca Valley Virus

SVV-001 Neotropix None 3 1 0 Neuroendocrine-featured tumours neuroblastoma and lung cancer

Coxsackievirus

Cavatak (CVA21)

Viralytics None 3 1 0 Melanoma breast cancer and prostate cancer

)5(ITCPWNQE[VGsOCETQRJCIGEQNQP[UVKOWNCVKPIHCEVQT05PQPUOCNNEGNNNWPIECPEGT4)ampTI)N[UROQVKH6-VJ[OKFKPGMKPCUG75WPKSWGshort 11 glycoprotein

REV IEWS

652 | SEPTEMBER 2015 | VOLUME 14 wwwnaturecomreviewsdrugdisc

copy 2015 Macmillan Publishers Limited All rights reserved