29
Viral gene therapy of “Multiple sclerosis”

Viral Gene Therapy ''Multiple Sclerosis'' by Farshid Mokhberi

Embed Size (px)

Citation preview

Viral gene therapy of “Multiple sclerosis”

Definition, Symptoms, Cuases: An autoimmune Inflammatory T cell dependent disease

that affect on myelin and distroy them in CNS.

Weakness in limbs, loss of centeral vision, double vision,tingling or pain in some parts of body, Electric-shock sensations that occur with certain head movements,Fatigue, Dizziness.

( all the symptoms are show up because of breakdown the communication.)

The cause of multiple sclerosis is unknown, some reserchear’s blivied in a combination of factors, ranging from genetics to childhood infections, may play a role.

Immune system:T helper

• Th1 cytokines are pivotal in the induction of human organ-specific autoimmune disease (ie Hashimoto's thyroiditis, Graves' disease, rheumatoid arthritis, insulin-dependent diabetes mellitus and MS) .

• Specialy Encephalitogenic T cell.

Important Risk Factors: Geoghraphic :

In both hemispheres, its prevalence increases with distance from the equator.

Ethnic:Different populations and ethnic groups have a markedly different prevalence of MS. The disease is especially common in Scotland, Scandinavia, and throughout northern Europe.

Genetics:First, second and third degree relatives of people with MS are at increased risk of developing the diseaseMS. Some scientists theorize that MS develops because a person is born with a genetic predisposition to react to some environmental agent.

Viruses: EBV, varicella zoster, Hepatitis vaccine

Perspectives in Gene Therapyfor MS:

Gene therapy for multiple sclerosis might include

different ‘human-grade’ vectors, which could be

used to deliver anti-inflammatory molecules as well as

neuroprotective agents into the CNS in a flexible and

useful way.

Therapies in Multiple Sclerosis:anti-inflammatory therapies:Deliver the ‘therapeutic’ molecules directly into the

CNS in order to inhibit blood-borne CNS-confined

Mononuclear cells acting as ultimate effector cells.

neuroprotective therapies:Rescue surviving oligodendrocytes.

Induce oligodendrocyte progenitor migration and

differentiation into demyelinating areas.

Anti-inflammatory Gene Therapies: Pro-inflammatory cytokines in MS:

Inf-Gama / (TNF-α/β)/ IL-2

downregulation of inflammatory mediators

Systemic delivery:

peripheral circulation encephalitogenic T cells.

CNS delivery:

directly into the CNS via the intraparenchymal or

intrathecal route.

Systemic Delivery:Experimental Autoimmune Encephalitis ( EAE) EAE

IL-4 / IL-10 / TGF-β 1

Autoreactive encephalitogenic T cells

Retriviral vector

Improved symptoms.

Note: opposite; TNF-α gene

Animal type:

mice and non-human primates (rhesus monkeys).

IL-6/ TNF- α/ IL-1b / IL-2 or IL10

Autoreactive encephalitogenic T cells

vaccinia-virus-derived vectors

( Adenovirus)

Improved symptoms.

Note: opposite; IL-4

Animal type:

mice and non-human primates (rhesus monkeys).

CNS Delivery:1) EAE

2) Co-stimulatory molecule CTLA-4-human immunoglobulin / IL-4

3) non-replicative adenoviral vector

infect : ependymal layer and leptomeningeal cells

produce a discrete amount of the transgene CSF for up to 1 month after injection.

improved EAE.

CNS Delivery continue: EAE IL-10/ IL-4 adenoviral vectors1) Delayed EAE onset2) Decrease in clinical score 3) Decrease in perivascular inflammatory infiltrates and in the

number of macrophages infiltrating the CNS parenchyma and the submeningeal spaces

4) Reduction in demyelinated areas and axonal loss.

No toxic reactions/ without any interference with the proper functioning of the immune system.improved EAE.

Animal type:Mice

neuroprotective therapies:1) transplantation of oligodendrocytes or o. precursors

Transplantation experiments :

• Defined demyelinating areas of experimental animals.

• transplantation is not a useful tool in multifocal CNS demyelinating diseases at present. ( MS)

2) Neurotrophic growth factors

• Migration/proliferation /differentiation

• Partially successful in EAE but not in MS patients.

3) promoting myelin restoration

Neurotrophic growth factors:1) EAE

2) fibroblast growth factor (FGF)-II

3) HSV-1-derived vectors

ameliorated EAE /without toxic reaction /inducing oligodendrocyte precursor proliferation and migration into demyelinating areas.

FGF-II was only seen when administered for up to 4 weeks, After that induced reactive astroglyosis.

Using FGF-II gene :CNS production of FGF-II (ependymal, choroidal and

leptomeningeal cells)

Decrease myelinotoxic cells (T cells and macrophages) both in the CNS parenchyma and in the leptomeningeal space.

Increas of the number of oligodendrocyte precursors and myelin-forming oligodendrocytes in areas of demyelination and axonal loss.

HSV-1 (d120) : Some of the limitations of the gene vector technology

so far employed in EAE/MS has been overcome by the

use of HSV-1-derived vectors :

• to be easily transferred within the CNS trough the CSF

Circulation.

• No major reactions of resident cells

• Intracellular machinery was used to produce the heterologous gene (eg cytokine genes).

Conclusions:Benefit:• Cytokine/growth factor gene therapy based on

injection of HSV-1-derived vectors into the CSF space might be a favourable approach for a chronic and multifocal CNS disease such as MS.

1. high cytokine/growth factor levels in all CNS.

2. therapeutic effect persists after a single vector

administration.

Our results indicate that intrathecal delivery of HSV-1-derived vectors containing anti-inflammatory cytokine genes may play a major role in the future therapeutic armamentarium of inflammatory CNS-confined demyelinating diseases

Problems: that rodent immunogene therapy cannot be transferred

easily to humans.

Encephalitogenic autoreactive T cells can be isolated from animal but in human the autoantigen(s) is still unknown.

vaccinia virus adenoviral vectors are immunogenic and they stimulate the immune sys and this feature makes it hard to use them.

HSV-1-derived vectors are short lasting and we should re-inject them.

QUESTION?

Thanks for your attention

References:

1.Kieseier BC, Storch MK,Archelos JJ, Martino G, HartungHP. Effector pathways inimmune mediated centralnervous system demyelination.2. Calabresi PA, Fields NS,Maloni HW, Hanham A,Carlino J, Moore J et al. Phase1 trial of transforming growthfactor beta 2 in chronicprogressive MS.3. The Lenercept Multiple SclerosisStudy Group and University ofBritish Columbia MS/MRIAnalysis Group. TNFneutralization in MS: results ofa randomized, placebocontrolledmulticenter study.

4.Martino G, Furlan R, Comi G,

Adorini L. The ependymal route

to the CNS: an emerging gene

therapy approach for MS.

5. Selmaj KW, Raine CS. Tumor

necrosis factor mediates myelinand

oligodendrocyte damage in

vitro.

6. Shaw MK, Lorens JB, Dhawan

A, DalCanto R, Tse HY, Tran AB

et al. Local delivery of

interleukin 4 by retrovirustransduced

T lymphocytes

ameliorates experimental

autoimmune encephalomyelitis.

7.Chen LZ, Hochwald GM,

Huang C, Dakin G, Tao H,

Cheng C et al. Gene therapy in

allergic encephalomyelitis using

myelin basic protein-specific

T cells engineered to express

latent transforming growth

factor-beta1.

8. Dal Canto RA, Shaw MK,

Nolan GP, Steinman L, Fathman

CG. Local delivery of TNF by

retrovirus-transduced

T lymphocytes exacerbates

experimental autoimmune

encephalomyelitis.

9. Croxford JL, O’Neill JK, Ali RR,

Browne K, Byrnes AP, Dallman

MJ et al. Local gene therapy

with CTLA4-immunoglobulin

fusion protein in experimental

allergic encephalomyelitis.

10. Krisky DM, Wolfe D, Goins WF,

Marconi PC, Ramakrishnan R,

Mata M et al. Development of herpes

simplex virus replication defective

multigene vectors for combination gene

therapy applications.

11. DeLuca NA, McCarthy AM,

Schaffer PA. Isolation and

characterization of deletion

mutants of herpes simplex virus

type 1 in the gene encoding

immediate-early regulatory

protein ICP4.

13.Kuklin NA, Daheshia M,

Marconi PC, Krisky DM, Rouse

RJ, Glorioso JC et al.

Modulation of mucosal and

systemic immunity by enteric

administration of non-replicating

herpes simplex virus expressing

cytokines.

14. Furlan R, Poliani PL, Galbiati F,

Bergami A, Grimaldi LM, Comi

G et al. Central nervous system

delivery of interleukin-4 by a

non-replicative herpes simplex

type 1 viral vector ameliorates

autoimmune demyelination.

15. Furlan R, Poliani PL, Marconi

PC, Bergami A, Ruffini F,

Adorini L et al. Central nervous

system gene therapy with

interleukin-4 inhibits progression

of ongoing relapsing–remitting

autoimmune encephalomyelitis

in Biozzi AB/H mice.

16. Poliani PL, Brok H, Furlan R,

Ruffini F, Bergami A, Desina G

et al. Delivery of a nonreplicative

herpes simplex type-

1 vector engineered with the

IL-4 gene to the central nervous

system protects rhesus monkeys

from hyperacute autoimmune

encephalomyelitis.

17. Ruffini F, Furlan R, Poliani PL,

Brambilla E, Marconi PC,

Bergami A et al. Fibroblast

growth factor-II gene therapy

reverts the clinical course and

the pathological signs of

chronic experimental

autoimmune encephalomyelitis

in C57BL/6 mice.

18. Mathisen PM, Yu M, Yin L,

Johnson JM, Kawczak JA,

Nishiyama A et al. Th2 T cells

expressing transgene PDGF-A

serve as vectors for gene

therapy in autoimmune

demyelinating disease.