101
Antiviral Research and Development Against Dengue Virus Bruno Canard, PhD. [email protected] 1

dengue

  • Upload
    ggggg

  • View
    172

  • Download
    0

Embed Size (px)

DESCRIPTION

dengue

Citation preview

Page 1: dengue

Antiviral Research and Development Against

Dengue Virus

Bruno Canard, PhD.

[email protected]

1

Page 2: dengue

Table of Contents

Part 1. Antivirals 3

A short historical view on antiviral research and therapies 3

Lessons learned from recent viral diseases and pandemies 3

The methods used to discover antivirals 4

Infected cell assays 5

Knowledge-based methods 5

The source of anti-infectious molecules 6

Why has natural product screening been neglected in antiviral research ? 8

Challenges associated with natural products in antiviral research 8

What is a validated antiviral target ? 9

Animal models 9

Patient cohorts and clinical trials 10

Frequent arguments about antiviral therapy feasibility 10

The introduction of dengue as a druggable disease 11

Diagnostics, and what does it tells us for antiviral therapy ? 11

Current treatment 11

Part 2. Dengue 13

Preamble 13

The Dengue Virus 13

The DENV targets for antiviral research 13

Overview of genome organisation 14

Overview of the DV particle and DV proteins as targets for drugs 14

The structural proteins 14

The Non-Structural proteins 15

RNA structures 17

The dengue validated targets 17

The cellular targets for antiviral research against dengue 18

siRNAs as tools and/or therapeutic agents 19

Response modifiers 20

Monoclonal antibodies 21

Mechanical devices 21

Part 3. Academic and academy-associated research centers 22

Part 4. The current industrial network of AV discovery 31

Part 5. Mapping the dengue drug design effort and needs 38

Annex 1. References 42

Annex 2. Patents 43

2

Page 3: dengue

Part 1. Antivirals

A short historical view on antiviral research and therapies

The first significant successes of anti-infectious disease treatments originated from the discovery and

use of antibiotics. The discovery of many viruses preceded largely the discovery of the first antiviral

molecule, which occurred at least 40 years after that of penicillin in 1928. The first documented

description of an antiviral molecule, that of 5-iodo-2'-desoxyuridine, occurred in 1959. It was

discovered active against Herpes ophthalmologic infections and followed by a series of related active

molecules. The fight against herpes was the perhaps the earliest and most significant driving force of

antiviral research. Herpes was the only significant viral disease for which all technical elements and

systems required to develop an antiviral molecule first became available (i.e., in vitro infected cell

systems, animal models, chronically infected patients,…). The antiviral drug field came of age in the

next decades with the first antiviral molecule finding its way to the clinic: Gertrude B. Elion

discovered acyclovir(2) a scientific breakthrough for which she was later awarded the Nobel prize in

1988. The subsequent emergence of AIDS in 1981, and the following pandemics drastically changed

the field of antiviral research, allowing the widening of concepts, technical developments, rules, and

business.

Lessons learned from recent viral diseases and pandemies

HIV and HCV: chronic invaders

The most important lesson comes from the following great achievement: it is possible to control a

chronic infection of a very sophisticated virus, such as HIV, that hides inside the chromosomes of the

infected cell. Although the victory is not total yet, it has profoundly changed the fate of the pandemic

victims, at least in western countries. After being inspired by other research fields, anti-HIV research

has “infected” other field of antiviral research and will continue to do so. Remarkably, after the

identification of HIV, the control of HIV through antiretrovirals originated from a collective effort on a

wide variety of scientific and medical fields, including efficient transfer from academia to the

corporate world. More recently, hepatitis C virus (HCV) research is now boosting the antiviral

chemotherapy field. Viral polymerases and proteases are targets par excellence, validated by the use

of inhibitors against HIV reverse transcriptase and protease, hepatitis B polymerase, and herpes virus

polymerase. Anti-HCV protease and polymerase inhibitors are in various stages of clinical trials.

Novel targets and cognate inhIbitors are adding to the list, such as the HIV integrase, and the HCV

NS5A.

HCV (genus Hepacivirus) and DENV (genus Flavivirus) belong to the same viral family Flaviviridae

sharing similar genome organization and replication strategies. Initially, research conducted on dengue

virus (DENV) was the actual starting and inspiration point for HCV research, when it became known

3

Page 4: dengue

that HCV had a flavivirus-like genome. Presently and conversely, knowledge and strategies gained

from the successful drug discovery and design process against HCV can now be translated back to the

DENV research field.

SARS and Influenza (H5N1 and H1N1): “hit and run” viruses

The SARS pandemic was due to a novel coronavirus which emerged in 2003 from China. The virus

took the world by surprise as coronaviruses were not known to cause life threatening pathologies.

Coronaviruses were clearly neglected viruses from the scientific and the medical/veterinary point-of-

view. The pandemic revealed blatantly our unpreparedness to such a problem: point-of-care in

hospitals crowded with contagious patients, high toll for clinicians, tracing secondary contacts of taxi

drivers and plane passengers, etc…The pessimistic say that nowadays viruses travel around the world

in 3 days. The optimistic say that social networks and cell phones make information travel much

faster. Perhaps the true challenge is elsewhere: making people believe and adhere to an “official”

information, as exemplified with the recent H1N1 crisis and the unsuccessful vaccination campaign. In

any case, this crisis has been the best advocate for antivirals as a complementary strategy to

prEvention and vaccination.

In the case of influenza, the size of the market has been the main booster of anti-influenza drug

development. This includes the availability of patients for clinical trials, and the fact that a potential

devastating pandemic would undoubtedly provoke stockpiling of antivirals in the time-window into

which an appropriate vaccine would available. Advice to stockpile anti-influenza drugs has been

recurrently advertised, mostly after 1995 when the 1918 spanish influenza strain genome was

published(8). well before the H5N1 and the H1N1 fear hit the world.

These two viruses do not produce chronic infections. These types of virus produce an infection

(unnoticed, mild, or acute) which resolves with virus clearance. This transient nature of the infection

has long been a problem to design an efficient therapeutic answer. Indeed, there are too many

unpredictable parameters to build a drug-design program based on traditional planning and funding

approaches. The two biggest problems are that it is impossible to evaluate precisely the market (and

invest accordingly), and that there is an unpredictable number of patients available for clinical trials.

The instructive aspect of these pandemics, however, is that they greatly contributed to re-shape

antiviral research at large (how can we anticipate? how money is going to be invested? ). These recent

crises have shaped considerably the grand public opinion towards the necessity to have broad-

spectrum anti-influenza drugs ready.

The methods used to discover antivirals

The original method of discovery of antivirals was partially a knowledge-based method, centered

around nucleobases and nucleosides (eg., uridine derivatives mentioned above against Herpes), known

to be used by viruses for their replication. The advent of AIDS and the discovery of non-nucleoside

4

Page 5: dengue

reverse transcriptase inhibitors opened the era of large-scale screening, which is entirely a trial-and-

error procedure, not based on previous knowledge. Millions of compounds are tested as fast as

possible (using high throughput screening (HTS) techniques), and only those showing activity are

selected.

Infected cell assays

In both cases (Herpes and AIDS), infected cell cultures provided the antiviral read out, before purified

targets were available and could be used. In these assays, compounds are tested individually to see if

they either cure an infected cell, or protect it from infection, pathogenic effects. The process is simple,

and relies on a cell culture system able to support virus growth. Not surprisingly, the discovery of

antivirals parallels the establishment of a robust infected cell based assay. When this was difficult or

even not possible (eg., HCV), the use of sub-genomic replicons or surrogate viruses has nevertheless

allowed drug discovery and design. There are now a wide variety of assay systems specific for each

virus. Robust dengue infected cell assays are available, highly efficient in terms of characterizing the

potency of a drug candidate. One significant disadvantage is the cost associated with cell culture

reagents and facilities, especially in low income countries. However, this method has an impressive

record of success compared to other methods.

Knowledge-based methods

The general trend is to reduce this trial-and-error approach and inject knowledge as much as possible

in the selection process so as to reduce costs and increase efficiency.

Computer-aided structure activity relationship (SAR) studies facilitate a responsive and efficient

management of research results and programs. Drug-resistance must be considered as part of the drug-

design process, as drug resistance mechanisms are being increasingly characterized and drug

combinations optimized, in order to avoid or delay resistance. The first large-scale effort to discover

anti-DENV drugs is to be credited to the Novartis Institute of Tropical Diseases (Singapore), who

conducted a complete screen of their proprietary chemical library against the DENV protease domain

from non-structural protein NS3 (see below).

Knowledge-based methods differ from classical cell based screening techniques in that they use

screening or discovery systems characterized at the molecular and sometime atomic levels. The

discovery system represents or approximates a given step of the virus life-cycle. The knowledge

associated with the system reduces the number of putative targets, and is supposed to provide directly

a mechanism of action of the compound or candidate drug. Examples of such systems are purified

enzymes used directly in the drug discovery test. It is expected that inhibition of the enzyme by a

compound in a test tube will mimic inhibition of the enzyme in the context of a viral infection. This is

5

Page 6: dengue

of course not granted. For a good enzyme inhibitor, the most frequent reasons of failure to inhibit a

virus in a cellular context are:

• The compound does not penetrate inside the cell.

• The compound is rapidly degraded/metabolized/transformed into an inactive compound

• The compound is toxic and of poor selectivity, ie., when used in a cellular context, the

compound will kill the infected cell and any direct effect on the virus is not apparent.

Many compounds can be selected as good inhibitors of a viral enzyme. However, the majority will fail

to convert into a candidate drug for the above reasons.

However, the main advantages of the method are:

• it discovers both a compound and its target at the same time.

• Currently, increasing general medicinal chemistry knowledge allows a better pre-screening of

compounds that have potential, ie., chemical libraries used as the source of molecules are each

day better in terms of containing “drug-like” molecules.

The future is the integration of both cell based assays and knowledge-based methods, to reduce the

time involved in i) finding the target at the molecular level, ii) having a trustable molecular/atomic

model to go quickly into hit-to-lead development by medicinal chemistry.

The source of anti-infectious molecules

Before their antiviral properties are discovered, antiviral chemicals or molecules either exist physically

somewhere in the world (and are selected or discovered), or they do not exist, and are invented and

subsequently synthesized. For molecules having a physical existence, they are either owned by

someone, and generally organized in a chemical library (or repository), or they are in the wild, in

plants, marine organisms, insects, etc…The issue of final ownership (ie., of a discovered molecule

having interesting properties) is then much more complicated.

This is an important distinction that has wide implications in drug discovery, from the ease of

discovery to the final ownership and availability to patients. The source of antiviral molecules is

indeed a key issue, particularly for dengue, for two main reasons:

• The cost of a drug is going to be a main issue because dengue occurs majoritarily in low

income countries.

• Although the low income dengue-afflicted countries generally do not have screening and drug

design facilities, most of the potential natural sources of drugs (mostly plants) are located in

these countries.

6

Page 7: dengue

Chemical libraries

The availability of large collections of pure compounds that can be handled, tested, analyzed and

whose compounds can be re-ordered has considerably evolved over the last decades. These large

collections were initially exclusively found in large pharmaceutical companies. Over the years, these

companies had accumulated compounds, assays, and know-how. The situation has drastically changed

over the last decade, mainly because robotics and bio-chemo-informatics have penetrated academic

modest-in-size labs and research structures. It has long been argued that screening was “an industrial

job” best accomplished in a corporate setting, an observation that was true to a certain extent, because

sophisticated robotics, engineering know how, and manpower was more easily mobilized there. The

decreasing cost of screening-associated technology, the diversity of the screening needs (targets,

pathways, organisms, pathologies,…), as well as the advent of proteomics and siRNAs (see below) has

done that many labs have their own screening facility, often small scale, for the defined process or

biological system they are studying. Likewise, many service centers and small companies are able to

propose screening as a service. Many large chemical libraries can be bought, several million pure

compounds are physically available to any purchaser. For a lab or company screening compounds

against a given virus or biological system, the problem is more to have original libraries. Indeed,

unique libraries that are not freely available minimize risks for a lab of being competed out, and

simplify the intellectual property of discovered molecules.

It looks that the tendency of screening very large collections of pure molecules is declining. This may

be due to the fact that methods to pre-screen virtually these collections have evolved to a point where

more focused libraries can be built, and small focused screens can be conducted on these “enriched”

libraires. Likewise, the increasing availability of atomic models (mostly crystal structures) of targets

make this preparation of enriched libraries much easier.

It is certainly too early to draw conclusions about the justification of great hype and faith on HTS

during the last two decades. There is a consensus to state that the number of drugs reaching the market

has decreased sharply in this period of time, relative to previous periods where drug discovery relied

much more on the screening and discovery of natural compounds (see below). The reasons are

certainly more complex than a mere wrong direction of the whole drug discovery and design world.

However, for a number of reasons, discussed below, the source of antiviral molecules will undoubtedly

evolve towards more screening from natural resources, blending with a great deal of experience in

high throughput techniques and medicinal chemistry expertise acquired in these past two decades.

Natural sources

Plants have been the traditional (and almost exclusive) source of active substances for most therapies.

At the present time plants are the indirect or direct source of ~ 50 % of approved drugs. Anticancer

drug research has been a leading force in natural product research and screening processes. From the

7

Page 8: dengue

1940s to 2007, 73% of the 155 small molecules approved as anticancer drugs were from natural

origin, directly derived from a natural product, or inspired by a natural product(4). To put antiviral

research in perspective, in the 1940s, there was not a single molecule known having an antiviral effect,

and the discovery and isolation of the first human viral pathogen was only 13 years away (Yellow

fever virus, in 1927). It is not surprising that the concepts of natural product screening, established for

cancer and inadequate for antiviral screening, had not entered the antiviral research field.

Why has natural product screening been neglected in antiviral research ?

First, many plant extracts are cytotoxic, a desired property for an anti-cancer drug. However, an

extract that kills the cell does not allow the monitoring of virus growth or inhibition. One has to use

extracts that are non-cytotoxic. By jeopardizing selectivity (ie, the ratio of inhibitory concentration for

the virus over toxic concentration for the cell), cytotoxicity has stopped many compounds or extracts

on their way towards antiviral pre-clinical trials. Second, when non cytotoxic crude extracts are used,

almost all of them exhibit antiviral activity. This antiviral activity is due mainly to compounds that

have no interest as antiviral drugs. These compounds are a wide variety of polymers, polyphenols, and

tannins. Third, if one avoids the above traps, screening natural extracts yields a lot of true inhibitory

molecules that are already known and characterized (and will fail in a composition-of-matter patent),

and have low potential for chemical modification into a useful and unique pharmacophore.

As consequence, during the past twenty years, the advent of combinatorial and parallel chemistry

coupled with high-throughput screening techniques has led to a decreased emphasis on plants (or

microbial, marine extracts) as a compound source. Nature has continued to inspire chemists and drug-

designers during the development of natural product-based compounds (such as antiviral nucleoside

analogues), but no natural product has actually been approved as an antiviral drug out of the 35 drugs

approved up to 2002. The trend seems to change as at least 8 natural products are since in clinical

trials in the field of virology (HIV and HCV), such as Calanolides A and B, DCK(PA-334B), 3,5-Di-

O-caffeoylquinic acid, MX-3253, 4-Methylumbelliferone, Bevirimat, Sho-shaiko-to H09, and

Sutherlandia frutescens(6).

Challenges associated with natural products in antiviral research

Whilst natural products as a source of drugs were falling out of favor of pharmaceutical companies,

the interest of this source was growing dramatically in countries were these resources are located, ie.,

mostly low income countries of the developing world. The main incentive was the adoption of the

Convention on Biological diversity, enforced in 1993. The challenges associated with this resource are

technical and policy issues(3).

On the technical side, the difficulty to deal with natural extracts has been developed above. There are

now an increasing number of methods reporting how to prepare an extract suitable to specific needs,

including antiviral research. Here also, technology has helped in the preparation of extract libraries

8

Page 9: dengue

pre-cleaned from unwanted substances described above (tannins, polymers, …). A second difficulty is

the variability of the source. Re-collection of the same plant may not give the same chemical

composition of an extract (different season, different development stage, misidentification, etc…). The

third difficulty is the resupply problem, particularly in large quantities. Over-harvesting may occur,

although it is sometimes possible to find alternate sources of the compound (plant cell culture, other

species, etc…). Last, isolation, re-synthesis of hemi-synthesis can be challenging, although science,

technology, and know-how are advancing faster than policy issues. However, natural products

collection and assays are located in the developing world, which is increasingly involved in finding

primary activities of an extract. When the next step in engaged, large pharmaceutical companies

having down-sized their natural product departments are not often ready to carry on. There is an

increase need to build intermediate/small dedicated structures in the corporate of academic world.

The policy issues associated with biodiversity exploitation address mainly the location of study of the

natural product collection. Authorizations, contracts and agreements can vary from extremely slow to

quite easy and diligent(3).

What is a validated antiviral target ?

A “validated target” is a cellular or viral component (protein, membrane, macroassembly,…) which,

when bound to a drug, leads to virus control (growth inhibition, elimination, virostatic,…) in the

infected cell, and hopefully, in patients eventually. Thus, any protein viral protein is not a validated

target, for example if, when inhibited or destroyed, the virus can still grow using and alternating

pathway compensating for the loss of its inactivated protein. Regarding the host cell proteins that can

be used as targets (ie., cellular proteins required for virus growth), blocking those proteins must be

both safe enough for the cell (ie., shown no toxicity at least for the uninfected cell) and the virus must

be unable to use an alternate protein compensating for the unavailability of the blocked protein.

Typical example are viral receptors. In many instances (eg., HIV), there are several cellular receptors

for the same virus. Blocking one receptor forces the virus to rely on other receptors, sometimes with

little effect on viral growth.

Animal models

In the case of dengue, the best animal model is the AG129 mouse, which has however a number of

drawbacks such as low and short viremia. Efforts are ongoing, mainly in academic labs to improve this

model. Surrogate systems exist that do not have these drawbacks (eg., the flavivirus Modoc virus),

and chimeric dengue/modoc viruses may yield interesting systems. Also, other animal systems are

being evaluated (golden hamster, macaques, …) with the increasing variety of dengue strains

available.

9

Page 10: dengue

Patient cohorts and clinical trials

The first large scale clinical trial has been conducted in Viet Nam (chloroquine), and another is

ongoing (see below, the Roche nucleoside analogue). This indicate that there is a sufficient and

sustained number of patients to go into clinical trials for any promising molecule. Other sites for

clinical trials will undoubtedly see the light, either in Asia or South America.

Frequent arguments about antiviral therapy feasibility

The most common argument opposed to antiviral therapy is that it would occur too late, ie., when the

viremia is already declining, low, or the virus cleared.

Diagnostic tests to rapidly detect DENV infection at an early stage (ie., early viremia) are currently

available (see below), and it has been demonstrated that there is a direct correlation between high viral

load and the development of the more severe, life-threatening form of dengue disease. The higher, the

worst. Thus, a drug reducing viral load at an early stage would potentially prevent DF and DHF/DSS.

Dengue viremia is short, being detectable only shortly before or concomitant to the onset of fever and

lasts four to five days after. The ability to rapidly diagnose dengue disease is thus key to the successful

implementation of antiviral chemotherapy.

Although virus may not be detectable in plasma, viral replication may be occurring in other cell

reservoirs, tissues, and body compartments where an antiviral drug could reach and target them. For

example, it is suspected that in addition to plasma leakage, the life-threatening DSS may involve

damage of organs such as the liver (DSS associated hepatitis) or heart (DSS-associated

myocardiopathologies).

In endemic outbreaks, prophylactic mass treatment around index cases would be essential. Rapid

diagnostics would detect infected yet asymptomatic people. Another as yet unevaluated consequence

of prophylactic treatment and decrease of viremia should appear in the vector-infection pattern.

Decreasing viremia in humans should result in a decrease in infected vector population and thus

impact on the transmission chain. Therefore, an efficient and safe drug, delivered early in the course of

dengue disease, should not only save lives but also curb potential epidemics.

An on-the-shelf drug allows a rapid response in the case of a sudden outbreak, and should not require

cold storage, an advantage for use in developing countries.

Lastly, in other deadly viral systems (Monkeypox virus as a surrogate system for Smallpox virus), 24h

post exposure prophylaxis with the drug cidofovir has been shown to significantly reduce mortality of

monkeys challenged with a lethal monkeypox infection, compared to 24h post exposure vaccination

which had no effect(7).

The second most common argument is that dengue occurs in low income countries where there is no

sizeable nor predictable market. Up to now, it is true that the common models of drug development

rested on analysis of an existing market, and financial planning and investment accordingly. It is

10

Page 11: dengue

apparent that this system will continue as it is for diseases that have a sizeable and predictable market,

most often chronic viral diseases. However, new business models have to appear for pandemic viruses

threatening many parts of the world simultaneously, irrespective to frontiers, the SARS –CoV and the

pandemic Flu being appropriate examples. In the case of dengue, the increasing incidence concerns

more and more countries, so a global response would be welcome. In addition, a potential dengue

drug would now cross the line of profitability. The number of companies interested in dengue

antivirals has increased sharply these past five years, and is still expected to increase.

It is also possible, at a very early stage in drug-design, to guide research on molecules that will be

cheap to produce. Cost-effectiveness is a crucial issue for this still poverty-linked disease. Cost-

effectiveness can be achieved by selecting as early as possible appropriate, easy to synthesize

chemical scaffolds, or by mining on active natural compounds (as discussed above) and selecting

those that represent major chemical constituents of appropriately available plants.

The introduction of dengue as a druggable disease

Dengue fever is not enough recognized as a major viral disease in terms of public health and economic

burden, although the situation is currently changing. The Jain PharmaBiotech report on “Antiviral

Therapeutics: Technologies, Companies, and Markets (February 2010)” mentions dengue within only

2 pages out of >450. However, the more aggressive expansion of the disease in the world, as well as

the emergence of a market, are attracting attention of novel actors in the field from both academic and

corporate world. There is no vaccine nor treatment available, and projections are that both will be

available approximately at the same time, ie., within ~5 years.

Diagnostics, and what does it tells us for antiviral therapy ?

There is no possible antiviral therapy without reliable diagnostics, and, in the case of dengue, rapid

diagnostics. In its early stages, dengue fever is often confused with other tropical diseases, which may

lead to inappropriate therapy. Since it is expected that cutting viremia as early as possible might result

in less severe dengue disease, dengue infection markers that parallel viremia are the best that one can

expect. This expectation is becoming reality with the presence and detection of dengue NS1 in

plasma/serum of patients though ELISA technology, preceding the appearance of dengue-reactive IgM

in the first days of illness.

Current treatment

The treatment is essentially supportive, and there is a very important challenge in estimating the

severity of the disease as early as possible. Indeed, unlike most other diseases, a key stage of the

disease is the patient’s defervescence during which increased vascular permeability may appear whilst

viral load and body temperature (fever) are declining. Close surveillance of this phase, which may

11

Page 12: dengue

need intravascular fluid replacement and maintenance of good haemodynamic stability, is crucial for

disease outcome.

Disease surveillance and characterization will certainly be key in the implementation of an effective

and appropriate antiviral therapy. Much is to be learned about targeting the virus in specific tissues

(liver, heart, brain,…) with appropriate antivirals in the future. The increasing knowledge about the

host response to a dengue infection is expected to bring most useful care and support to dengue

patients, whereas knowledge of DENV components and life-cycle will eventually lead to efficient

antivirals, most probably in combination with those targeting cellular targets (see section “The cellular

targets for antiviral research against dengue”).

12

Page 13: dengue

Part 2. Dengue

Preamble

There are a number of recent high quality scientific reviews about dengue, the DENV, and anti-dengue

drug design. Amongst the most complete and relevant are:

• Dengue (2008) Tropical Medicine: Science and Practice, Vol. 5, Scott B. Halstead Editor,

Imperial College Press.

• Gubler, D., Kuno, G., and Markoff, L. (2007) Flaviviruses, in Fields Virology, Vol.1 Fifth

edition, LWW, Knipe DM and Howley PM, Eds.

• Noble, C. G., Chen, Y. L., Dong, H., Gu, F., Lim, S. P., Schul, W., Wang, Q. Y., Shi, P. Y.

(2010) Stategies for the development of DENV inhibitors, Antiviral Research, 85, 450-462.

The reader will find a lot of references in these documents. For the sake of clarity, only key references

and references that may not be in these documents will be cited in the following text. Much of the

work has been performed in the laboratories mentioned in the academic labs and center list below.

Lastly, the list of patents sorted by year in annex 1 is also a valuable source of information.

It is not the intention of this document to re-formulate what has been excellently written by expert

colleagues in the field. Rather, the purpose of this document is to connect many known or emerging

facts about anti-dengue drug design, evaluate where we presently are, what and where are the needs

and challenges to promote and sustain an active anti-viral drug design field.

The Dengue Virus

The DENV targets for antiviral research

Flavivirus and Flaviviridae research has led to the characterization of an increasing number of viral

encoded proteins and enzymes, including envelope and capsid proteins, polymerases, helicases and

proteases. Processes involved in the entry of DENV into cells (virus-receptor binding, E protein

conformational changes, virus internalisation and membrane fusion) are being more and more

understood at the molecular level. For DENV whose RNA genome is decorated by a type-1 cap

structure, enzymes involved in cap formation such as the RNA triphosphatase, guanylyltransferase

(still unknown) and methyltransferase are additional potential targets. Considerable progress has been

made in their characterization. Chemical libraries from natural and synthetic origins can now be

screened against these novel pathways and targets.

13

Page 14: dengue

Overview of genome organisation

The genome organisation is that of single positive strand RNA genome virus, ie., it is similar to a large

cellular mRNA molecule. The genome is approximately 11 kb in size, bears a type I cap structure at its

5’-end, and lacks a 3’-polyadenylate tail. The long open reading frame encoding a large polyprotein is

flanked in 5’ and 3’ by untranslated regions (UTRs). The latter carry a number of cis-acting signals

(stem loops, conserved sequences, …) required for viral replication, and possibly RNA capping. There

are complementary sequences in these UTRs that are thought to be responsible for cyclization of the

genome, which is essential for replication.

Overview of the DV particle and DV proteins as targets for drugs

Flavivirus are enveloped viruses having two outer membrane proteins, the envelope (E) and the

membrane (M) processed from the precursor prM. The genome is thought to be wrapped/associated

with the capsid protein C. A single polyprotein is translated from the genome, and the former is

cleaved by a combination of cellular proteases and a viral serine protease made of NS2B and NS3

(protease N-terminus domain).

Figure 1. The DENV (+)RNA genome and it co-linear polyprotein.

Proteolysis yields ten proteins, the three structural proteins (C, prM, and E) and the seven

nonstructural (NS) proteins involved in genome replication and capping (NS1, NS2A, NS2B, NS3,

NS4A, NS4B, and NS5). Some of these NS proteins also participate in pathogenesis and counteract

the innate immunity of the host cell.

Replication of the viral genome does not occur freely in the cytoplasm. Instead, there is an extensive

intracellular membrane re-arrangement in the infected cell, with various observable cell substructures

containing most NS proteins organized along the virus replication cycle.

The structural proteins

The M and E proteins have been considered so far as drug targets. The E protein is endowed with a

dual function : to recognize cellular receptor, and to fuse the viral membrane to cellular endosomic

membranes. Five receptors have been found to be involved in binding to E : DC-SIGN, L-SIGN, the

high affinity laminin receptor, the mannose receptor, and GRP78. E is an attractive target because

theoretically, an antiviral molecule binding and impeding attachement would act before infection or

spreading the virus to yet-uninfected cells. In the RNA virus antiviral world, there are several

examples of such inhibitors targeting early phase of the viral life cycle : that of T20 (a fusion inhibitor

14

Page 15: dengue

corresponding to the C terminus ectodomain of gp41) in anti-HIV therapy(1) and that of Pleconaryl, a

small molecule enterovirus capsid binder(5).

Several disadvantages are associated with these drugs : peptide inhibitors are delivered intravenously,

and picornaviral capsid binder elicits quickly drug resistance. Parenteral delivery is a serious

drawback for any dengue drug which would preferably be delivered with limited hospitalization and

epidemic settings, and drug resistance might not be such a crucial issue as it is for chronic viral

infections.

The dengue E protein crystal structure is known. E belongs to class II fusion proteins. Upon binding to

a receptor and endocytosis under a trimeric form, the acidic environment of the endosome induces a

structural re-arrangement yielding fusion of cellular and viral membranes. Theoretically, a similar

strategy as that of HIV and T20 could be followed. Crystal structure study of a E fragment has

revealed a pocket that could be used for antiviral drug-design. The crystal structure of the prM protein

bound to E (prM-E heterodimer) is known, opening avenues for drug design. The structure of the

capsid protein C has bee elucidated in solution. The E protein is the most obvious target for

therapeutic monoclonal antibodies (see below).

The Non-Structural proteins

Out of the seven NS1-to-NS5, only NS3 and NS5 have been considered so far as drug targets, not only

because they are essential to virus growth but also because they exhibit enzyme activity, which is a

plus regarding drug screening. The role and structure of NS1 is unknown. It is a soluble protein

detected very early during infection, but has received little attention so far as an antiviral target. NS2a,

NS2b, NS4a, and NS4B are membrane –associated proteins believed to anchor and regulate the

replication complex during the virus life-cycle.

• NS3 (69 kDa) carries two functional domains, a N-terminal serine protease(~170 aa), and a C-

terminus helicase/RNA triphosphatase (~440 aa). The protease domain is inactive alone, and

needs the presence of 40 aa of NS2b bound to form a protease active site. The NS2b/NS3

protease has been the first dengue protein target actively used in drug design programs.

Tragically, the crystal structure reported in 1999 for this domain was fraudulent, and the

original article retracted in 2009. Two complementary approaches have been followed to

discover antivirals based on the inhibition of this enzyme. The first approach has been the

screening of a large chemical library (1.4 million compounds by NITD alone, see on the list of

companies below) and the second approach has been to design peptidomimetics, an approach

which has also been followed in the case of HCV. So far, the relatively flat topology and the

charge repartition of the NS2b/NS3 protease active site is believed to account for the difficulty

of finding potent compounds. However, such difficulties have also been mentioned many

times for the HCV protease, but patience and obstination have finally payed off with the

15

Page 16: dengue

discovery of potent HCV NS3 protease inhibitors.ne other potential problem is that the

protease domain might be regulated by its C-terminus fellow helicase domain, as different

conformations of the full-length NS3 have been reported. A great deal of knowledge has been

accumulated and published on this protease, and progress are still accumulating in this field,

so the future may be more favorable, perhaps in combination with other dengue drug/target

pairs. The NS3 helicase domain is also an interesting target because it contains features unique

to flaviviruses, such as Domain III. However, drug discovery and design against this enzyme

has proven challenging for several reasons such as poor helicase activity in vitro, a too-open

ATPase active site, and the absence of obvious pockets able to accomodate small molecule

inhibitors. Several HTS assays have nevertheless been developed, but so far, no convincing

small molecule inhibitor has been reported, a situation paralleling that of HCV helicase.

• NS5 is the largest and most conserved and most conserved dengue protein. NS5 is a 900

amino acids protein (~100 kDa) carrying the enzymatic activities required for RNA capping

and synthesis of the dengue RNA genome. The NS5 N-terminal domain has been shown in

2002 to be a 2’O Methyltransferase (MTase) through crystal structure analysis, and later, the

N7-guanine MTase activity was also demonstrated to be embedded in the 260 amino acid

fragment. The NS5 C-terminus domain has been shown to carry RdRp activity in in vitro

assays, and its crystal structure has been determined in 2007 simultaneously to that of West

Nile virus. The full structural picture will be completed when the full-length NS5 crystal

structure is going to be available. In a parallel to HIV and HCV drug design programs, the

knowledge of a structure of a ternary complex made of NS5/RNA/NTP would certainly add

excitement to this growing active field. Indeed, « naked » polymerase structure used in drug

discovery often point to inhibitor compounds that are sub-optimal when assayed against

replication complexes. Two main types of inhibitors have been described for polymerases.

The nucleoside analogues are substrate mimics that, once activated and incorporated into the growing

RNA chain, stop RNA synthesis, hence they are called « chain terminators ». To do so, nucleosides

must be phosphorylated by host cell kinases up to the 5’-triphosphate state, then compete with natural

substrate selectively at the viral RNA polymerase active site. This concept has met with impressive

success in the case of HIV and other viral DNA polymerases. In that case, ie. DNA polymerases, the

5’-triphosphate nucleoside analogue is competing with micromolars of natural dNTP substrates. In the

case of viral RNA polymerases, the problem is complicated by the fact that analogues have to compete

with intracellular millimolar concentrations of NTPs. Large scale screening of these analogues relies

on using libraries focused on the nucleoside motif. Because of the required activation through 5’-

phosphorylation, screening can only be performed on infected cell cultures.

16

Page 17: dengue

High throughput screening methods have introduced the novel concept of non- nucleoside analogues,

ie. random chemical structures of small organic molecules able to bind specifically to a viral target.

The major challenge for these inhibitor ligands is to target a conserved pocket not so prone to

mutation, otherwise, drug resistance will occur very quickly though rapid appearance of mutations.

For dengue, it remains to be evaluated whether or not drug resistance is going to be a significant

problem. Or these compounds, initial large scale screening can be achieved using subgenomic

replicons, infected cells, and purified enzymes (see above « the methods to discover antivirals »).

NS5 may also play a role in pathogenesis, so its targeting may hit two birds with a single stone. NS5

has been reported to interact with STAT2, perturbate interferon signaling, and its traffic within the

nucleus of the infected cell is far from being understood.

RNA structures

The dengue genome is a single stranded RNA molecule of positive polarity. However, the replicative

form of dengue RNA is not a single linear molecule but rather, a cyclic or dimerized genome. This

special genomic RNA organization proficient for replication carries many highly ordered secondary

and tertiary structures ensuring proper regulation of dengue RNA synthesis. Most of these RNA

structures are located in the 5’ and 3’ untranslated regions. For example, the TIA-1 and TIAR antigens

have been identify to interact with 3’-stem loop structures and inhibition of their interaction has an

antiviral effect in infected cells (see patent list). Phosphorodiamidate-linked morpholino

oligonucleotides (PMOs) have also been shown to target efficient RNA stem-loop structures (see

patent list, 2007). Since they are unique to the viral RNA genome, and since the scientific field of

small RNAs is booming, it is almost certain that these RNA regions contain a significant potential for

drug discovery and design, yet to be addressed and validated.

The dengue validated targets

Presently, there are no drugs against dengue in the clinic. Therefore, what we call a validated target is

only derived by analogy to other viruses for which drugs have proven to be effective. Herpes and HIV

have been the drug-design founding viruses. Now for DENV, HCV is fulfilling this role. Recent data

presented regarding a nucleoside analogue, although toxic, have shown that the NS5 protein is a

validated target (see compound NITD008). The validated targets are thus the RNA-dependent RNA

polymerase and, by analogy to HCV, the protease. More recently, the HCV NS5A protein seems to

emerge as a very interesting target, but NS5A does not have an equivalent in dengue, so far.

Conversely, the dengue RNA genome is capped, and the DENV genome encodes most of its own RNA

capping machinery. This is not the case for HCV, which does not rely on RNA capping for gene

expression. The RNA capping enzymes of dengue so far await validation in an animal model both at

the level of efficiency, and toxicity. Indeed, it is not known if the abundance of cellular MTases will

17

Page 18: dengue

cause a specificity problem, ie., if it will be possible to design an anti-dengue MTase inhibitor having

non-significant toxicity effect through co-lateral inhibition of host cell MTases.

The cellular targets for antiviral research against dengue

The host cell is actively involved at many levels during DENV infection, either at the level of innate

immunity and counteraction thereof, or providing co-factors and template for replication of the virus.

In theory, any of the cellular proteins involved in DENV life cycle is a potential target for antiviral

therapy. The strategy may differ if the cellular protein has to be activated or inactivated. Several

proteins belong to the former case, such as RNase L involved in innate immunity. In the latter case, the

protein might be actually used by the virus to promote its own growth, and the cellular protein will

have to be inhibited or this promoting activity inactivated somehow. As an example, this could be the

case for furin-like proteases and signal peptidases initiating the dengue polyprotein processing. A third

category of cellular targets is that of cellular proteins involved in pathogenesis and not viral replication

directly. These proteins certainly represent interesting targets, but presumably, an antiviral drug effect

would have to be fine-tuned to avoid complete repression of these host defense factors, yet conserve a

sufficient effect to dampen the excessive host response responsible for pathogenicity.

In any case, the two main pre-requisites for host factor inhibitors are that the cellular protein or

macromolecule can effectively be used as a template for drug design (ie., acceptable druggability) and

that, when used, there is a non-significant or acceptable level of cytotoxicity.

There are a number of known cellular proteins and pathways that exert an anti-dengue effect when

affected or inhibited. Proteases and glucosidases constitute the earliest discoveries of such host

factors, whereas other candidates (kinases, cholesterol synthesis enzymes, proteins involved in

immune response,…) are progressively discovered and validated through siRNA studies (see below).

Cellular proteases

They are mainly of the furin and signal peptidase type. Furin is involved in the maturation of the M

protein from its precursor prM encoded in the dengue polyprotein. The consensus sequence cleaved by

furin is well defined and could theoretically serve to design inhibitors. However, the delicate

specificity balance between host cell targets of furin and dengue prM has not been fully investigated,

and it is not known at this stage if it would be achievable without side effects. The second candidates

are the signal peptidases, located in the ER membranes, which initiate further dengue polyprotein

processing before NS2b/NS3 protease takes over and maturates the whole NS enzymes. As in the case

of furin, the specificity and balance of effect has not been fully evaluated.

Glucosidases

Several DENV proteins (prM, E, and NS1) are decorated by glycosylation upon travelling through the

ER. They are however further maturated upon de-glycosylation by cellular glucosidases I and II,

18

Page 19: dengue

which leaves a single carbohydrate unit at their surface. It has been shown that inhibition of these

enzymes has a potent antiviral effect, since these maturation events are required for proper folding of

the viral proteins. Glucosidases have a very long record of study regarding their inhibition, and many

carbohydrates and carbohydrate mimics have been synthesized and shown to be potent glucosidase

inhibitors in vitro and in vivo. Castanospermin and deoxynojirimycin derivatives have been evaluated

against dengue (and other viruses) and shown interesting antiviral effects. Castanospermin seems

specific for dengue when assayed on West Nile and Yellow Fever viruses. Interestingly, the compound

is safe in mice and protects them efficiently against lethal DENV challenge, indicating that this

research avenue is worth further effort.

Other recent targets

A screen of 120 kinase inhibitors resulted in the discovery of the anti-dengue effect of dasatinib, a

known c-Src kinase inhibitor. Phosphorylation of proteins by kinases is involved in many signal

transduction and regulation pathways such as endocytosis, cell survival and immune evasion during

viral infection. It thus represents a potential avenue to design potent anti-dengue drugs since the kinase

inhibitor field is very active and has produced a very large number of original compounds. Another

recent point-of-action for the dengue drug designer is the cholesterol metabolism, since membrane

cholesterol is involved in DENV (and other flavis) entry and replication. The specific targetting of

cholesterol synthesizing enzymes through siRNA has shown that this avenue is promising, and again,

since cholesterol metabolism is an active area in drug design, crossover drugs may unexpectedly

appear as the dengue problem reaches the interest of mainstream pharmaceutical companies.

There are a large number of viral-cellular protein interactions that have to be discovered and that will

constitute targets to either control viral growth or pathogenicity, or both. A significant dengue-cellular

protein interactome is not known yet, but few discoveries in this direction have already provided

potentially interesting interesting avenues. For example, the binding of STAT2 to DENV NS5 and the

resulting dampening of the interferon response illustrates a direct interaction at work, whereas the

impact of importin a and b binding to the NS5 polymerase has not been fully evaluated yet. Many

protein-protein interactions are to be discovered and will provide novel drug design subjects, alone or

in conjunction with other targets.

siRNAs as tools and/or therapeutic agents

Recently, genome-wide studies have allowed a complete re-assessment of host factors involved in

dengue infection. The use of large-scale siRNA screens has generated large list (>100 proteins) of host

factors involved in helping DENV to achieve its replication cycle. These host factors are potential

targets for drug design taking into accounts the caveats mentioned above. It is interesting to note that

very few of the previously known host factors (proteases, glucosidases, etc…) have been found with

these siRNA screens, indicating that a better resolutive power of these techniques is expected to yield

19

Page 20: dengue

additional targets. Also, these pioneering studies aim at monogenic effects, and future screens will

certainly address more precisely the identification of several genes acting inside a pathway requested

for DENV growth. In addition, these screens did not (in fact, could not) identify innate immunity

genes that defend the cell against DENV infection. The way these defenders are induced and regulated

will certainly provide interesting avenues of research in the future.

Are siRNA interesting as therapeutic agents per se? siRNA have largely proven their efficacy in vitro,

but there are several hurdles that have to be overcome before they become drugs. The most important

issue is the delivery of siRNA in patients. Since tissue tropism is a key issue in viral infections, the

only almost certain use of siRNAs is for delivery in the skin or in the liver, which for dengue disease,

is not sufficient yet. siRNAs will be either delivered (synthetic modified siRNAs of increased

stability) or made available through in vivo expression, the latter being far from reaching anti-dengue

clinical application.

Response modifiers

As any pathogenic virus, DENV infection provokes a large number of distincts and intertwined

responses in the host cell, tissues, and whole organism. Whilst it is probably not necessary to engage

into a therapeutic action against a mere fever, there has been considerable interest in monitoring signs

that would predict the potential evolution of the patient towards severe disease forms. Indeed, after

initial fever and detectable viremia, there is a partial remission generally followed by severe effects

that can lead to fatalities though hemorrhagic fever and shock syndrome. Therefore, any treatment that

would dampen or control this dangerous secondary response without jeopardizing the host antiviral

response would certainly find its place in the therapeutic arsenal.

The most common observation around hemorrhagic fever and vascular leakage, are

thrombocytopoenia, neutropoenia, elevated liver enzymes, elevated serum cytokine levels and

disseminated intravascular coagulation. Hemorrhagic fever can progress to a life-threatening shock

syndrome, a hypotensive state with unrecordable pulse and blood pressure.

A lot of current research is ongoing on the pathogenesis. The pathogenic effects of this secondary

response are believed to be due to a “cytokine tsunami” involving T cells types: Th1 switching to Th2,

TS, Th3, Tr1, memory T, and TCF, on top of which B cell response and antibody dependent

enhancement may synergize the adverse effects.

During the last decades, nitric oxyde induction has been pointed as been associated to many virus

infections. NO level changes have been observed upon dengue infection of cells, mice, and patients.

The role of NO has been investigated, but its pleiotropic role in general metabolism, immune system,

and inflammation, precludes a clear answer regarding its involvement in pathogenesis. Precise control

of NO synthase(s) remains to be experimentally achieved to decipher the precise NO role according to

patient (eg., previous infection), virus serotype, and other parameters. It may then well be that these

data would serve to adjust the host response to desired levels. NO has be reported to directly inhibit

20

Page 21: dengue

the DENV RNA-dependent RNA polymerase, and a potential site of action has been mapped onto NS5

which could explain the different sequence-dependent NO sensivities observed. Again, this

preliminary data deserves a closer look on larger samples of patients and viruses.

Monoclonal antibodies

Many different monoclonal antibodies have been raised against dengue proteins. Although it seems

that the cost associated with both production and use remain prohibitive, only the future will tell if this

therapeutic avenue becomes available cost-effectively in clinical settings.

From the scientific and medical point-of-view, the first most important problem is to address the

Antibody Dependent Enhancement (ADE) of infection problem upon use of an antibody. This problem

is common to Mab and vaccine design. Several antibodies have been described, and in most cases,

their therapeutic potential has been examined in the context of the ADE problem. Not surprisingly, all

of them are directed against the envelope protein. Antibody engineering to prevent FcγR binding

shows potential to design safe and potent therapeutic antibodies. Several pharmaceutical companies

involved in this research avenue are listed below.

Mechanical devices

Mechanical devices have been proposed to assist in the treatment of drug- and vaccine-resistant

pathogens. These devices are to be used during the viremic phase during which circulating viruses are

trapped by the device fed by the blood stream. Purified blood is produced and re-delivered to the

patient, and the treatment is sought to provide first line countermeasure in the absence of drug or

vaccine treatments. The technology converges the blood filtration principles established in

hemodialysis and plasmapheresis with the immobilization of affinity agents (eg., lectins) that capture

enveloped viruses by the surface carbohydrate structures they have evolved to evade the natural

immune response. The device increases the likelihood that a patient's own immune response can

overcome infection (see patent list 2009). It is not yet known if such devices can effectively achieve a

sufficient drop of virus titer to prevent severe dengue.

21

Page 22: dengue

Part 3. Academic and academy-associated research centers

Support of dengue antiviral research

With the increasing awareness of dengue emergence and spread around the world, support for dengue

research has been growing, too. Many countries have government agencies funding dengue research in

specific or general programs. The most active (ie., having a significant number of projects funded in

the last 10 years) and large-scale funding agencies are the National Institute of Health (USA), the

European Research Area (EU, Framework programs 4-7), and the Wellcome Trust (UK).

• National Institute of Health (NIH, through its own National Institute of Allergy and Infec-

tious Diseases (NIAID), is currently supporting many projects on dengue antiviral develop-

ment. Several efforts are currently ongoing to develop drugs that target viral and cellular pro-

teins required for dengue replication and therapeutic monoclonal antibodies. NIAID is also

supporting the screening of compounds for efficacy in vitro against dengue. Several thousand

compounds have been screened since 2004 and approximately 50 of them have been identified

for further evaluation. In addition, NIAID is supporting the development of new murine mo-

dels for dengue. These models will be used to evaluate the in vivo efficacy of promising com-

pounds submitted by the scientific community. A list of projects currently funded by NIH on

dengue and other diseases can be found at: projectreporter.nih.gov/

• European Research Area, The European commission has been supporting research programs

on infectious diseases and other since 199’ in its 4th framework program (FP), up to its its 7 th

ongoing FP. A total of 25 projects on dengue have been supported, which can be found at the

following url: http://cordis.europa.eu/search/index.cfm?

fuseaction=search.resultlist&#q=27AB3D69F84955931621A4395513A8D9@showtype=proj

@page=1@perPage=10@sortBy=RELEVANCE@sortOrder=DESC

The most recent large-scale funding on dengue antiviral drug discovery and design is from the

FP7-SILVER project, which has dengue as one out of its three main targets for antivirals.

• The Wellcome Trust, The Wellcome trust has been supporting many projects on dengue over

the past 10 years (see http://www.wellcome.ac.uk/). In 2009, this institution has funded prob-

ably the most important funded dengue research project entirely dedicated to drug design

(http://www.wellcome.ac.uk/News/2009/News/WTX057192.htm) with a total of 2.6 million €

for the whole project.

22

Page 23: dengue

• The International Consortium on Anti-virals (ICAV), The International Consortium on An-

ti-Virals (ICAV, see www.icav-citav.ca) is a not-for-profit drug development organization ded-

icated to the discovery and development of antiviral therapies for neglected and emerging dis-

eases. ICAV operates through the establishment of network of collaborative labs on defined

viruses and viral targets. ICAV raises awareness of governements, funding agencies and di-

verse stakeholders about possible pandemies and emerging virus threats. Dengue has been and

is on their priority list. ICAV has a potential of rapid mobilization of a defined lab network to-

wards solving a problem related to the development of any antiviral (eg., hit discovery, ADME

tox studies, etc…). One long term goal of ICAV is global access to affordable anti-viral ther-

apies.

23

Page 24: dengue

Academic and Academy associated research centers

• Center for Infectious Disease Research, University of Queensland, Australia

(http://www.cidr.uq.edu.au) The CIDR has long been involved in dengue research and more

specifically into antiviral drug target characterization, drug discovery and design, with at least

5 of it research groups having dengue and flaviviruses as a major area of study. Small

molecule library screening has identified E protein ligands with anti-flavivirus properties.

Specific research teams: Alexander Khromykh, RNA Virology Laboratory, Dr. Paul Young,

and and Roy Hall, Molecular and Virology Unit

http://www.cidr.uq.edu.au/?page_id=46

http://www.cidr.uq.edu.au/?page_id=76

http://www..cidr.uq.edu.au/?page_id=122

• Drexell University, PA, USA (http://www.drexel.edu) Drexell University (Drexel Institute for

Biotechnology and Virology Research), in association with PharmBridge, has drawn on the

known effect of cellular glucosidase inhibitors, and synthesized novel series of compounds

around the iminosugar pharmacophore. Pharmabridge Inc. (Pensylvania) has no dedicated

Dengue research per se but provides contract research and development activities for

pharmaceutical, biotechnology and agricultural industries. Drexel teams have recently

reported the broad spectrum anti-dengue activity of alkylated porphyrins, geneticin, as well as

that of alpha glucosidase inhibitors in combination with ribavirin. Specific research team:

Microbiology and Immunology, Drexel Institute for Biotechnology and Virology Research,

Dr. Jinhong Chang, http://www.drexelmed.edu/Home/AboutOurFaculty/JinhongChang.aspx

• DUKE-NUS Graduate Medical School, (http://www.duke-nus.edu.sg ) Program in Emerging

Infectious Diseases, Singapore, has been performing genome-wide analysis to identify host

factors involved in dengue infection as well as investigating monoclonal antibodies NS3

potentially interesting in therapy. Specific research teams are hosted in the department of

Emerging Infectious Diseases, whose program director is Dr. Gubler, Duane J.,

http://www.duke-nus.edu.sg/web/research_signature_research_programs_emerging.htm

• Florida Gulf Coast University, Department of Biological Sciences, Fort Myers, Florida,

USA, (http://www.fgcu.edu/cas/Departments/biosci.html) has identified p-sulfoxy cinnamic

acid as an entry inhibitor, and also patented peptides targeting the entry process of dengue

24

Page 25: dengue

virus. Specific research team: Biotechnology Research Group, Drs. Scott F. Michael, Sharon

Isern, and Joshua M. Costin , http://www.fgcu.edu/CAS/Biotechnology/ALC.html

• Fundación Instituto Leloir, Buenos Aires, Argentina, (http://www.leloir.org.ar) has

characterized promotor and cyclization elements of the dengue RNA genome that bear

potential as original antiviral targets. Specific research team: Molecular Virology Laboratory,

Dr. Andrea Gamarnik

http://www.friendsofleloir.org/research/molecularvir.htm

http://www.hhmi.org/research/international/gamarnik_bio.html

• The Harvard Medical School, Department of Biological Chemistry and Molecular

Pharmacology, Boston, Massachusetts, USA, (http://bcmp.med.harvard.edu/) has produced

pioneering information on the E protein structure and mechanism, identified a pocket suitable

for ligand binding, and followed peptide design to inhibit the dengue virus entry/fusion

process. Specific research team: Laboratory of Structural Cell Biology, Dr. Stephen C.

Harrison

http://crystal.harvard.edu

https://bcmp.med.harvard.edu/faculty/harrison

• Institut Pasteur -Hong Kong (http://www.hkupasteur.hku.hk) has developed and patented a

cell-based system to produce virus-like particles for the four serotypes of the dengue virus by

co-expressing the two prM and E viral structural proteins in host cells. Applications are

foreseen in serodiagnosis, monoclonal antibody development, drug screening and vaccine

development. Specific research team: Virus-Host Interactions, Dr. Béatrice Nal

http://www.hkupasteur.hku.hk/index.php/research/virus_cell_biology

• Institute for Antiviral Research, Utah State university, Logan, USA

(http://www.usu.edu/iar/iar_home.html)The Institute for Antiviral Research of Utah State

University performs research oriented toward the control of viral diseases. Researchers of the

Institute perform fee-for-service evaluations for corporate industry. Evaluation of 2’-C methyl

nucleoside, initially intended for HCV, has been evaluated for dengue. Specific research

team: Dr. Justin Julander, http://www.usu.edu/iar/Faculty/justin/justin_home.html

• Instituto di Ricerca in Biomedicina, (http://www.irb.ch), Bellizona, Switzerland is

investigating antibodies and their therapeutic potential against dengue, studied amongst other

25

Page 26: dengue

viruses. Developments on dengue are collaborative with HuMabs, a biotech company

belonging to a company consortium operated by Synergenics LLC

http://www.synergenics.net. Specific research teams: Cellular immunology,

Dr. Federica Sallusto,

http://www.irb.ch/index.php?

option=com_jresearch&view=researcharea&task=show&id=3&Itemid=86,

Immune regulation, Dr. Andrea Lanzavecchia,

http://www.irb.ch/index.php?

option=com_jresearch&view=researcharea&id=2&task=show&Itemid=112

• John Paul II Catholic University of Lublin, Department of Molecular Biology, Lublin,

Poland (http://www.kul.pl), has synthesized inhibitors of the Flaviviridae helicases, peptide-

like and small molecules. A collaboration is active with the Laboratory for Drug Design and

Synthesis, University of Maryland, Baltimore, Maryland, USA. Specific research team:

Department of Molecular Biology, Dr. Borowski Peter,

• Mahidol University, Laboratory of Molecular Virology, Institute of Molecular Biosciences,

Nakornpathom, Thailand, (http://www.mb.mahidol.ac.th/mben) has been conduction structure-

base drug design against the protease NS2b/NS3. The National Center for Genetic

Engineering and Biotechnology (BIOTEC), National Science and Technology Development,

Mahidol University (www.biotec.or.th/biotechnology-en/en/index.asp) has discovered

Viprolaxikine, a small cytokine-like protein with strong anti-dengue effects in dengue virus

infected cells. Specific research team: Virology Research Group, Dr. Duncan R. Smith

http://www.mb.mahidol.ac.th/mben/index.php/research/research-group/virology

• Memorial Sloan-Kettering Cancer Center, HTS Core Facility, Molecular Pharmacology

and Chemistry Program, New York, USA (http://www.mskcc.org/mskcc/html/52147.cfm).

The center has established screening facilities and know-how, and recently reported the

screening of 5,632 well-characterized bioactives, yielding 73 confirmed compounds with

IC₅₀ potencies ranging from 60 nM to 9 μM and yielding a hit rate of 1.3%. Specific

research team: HTS Core Facility, Molecular Pharmacology and Chemistry Program, Dr.

Hakim Djaballah

http://www.mskcc.org/mskcc/html/52402.cfm

http://www.mskcc.org/mskcc/html/5785.cfm

26

Page 27: dengue

• Nanyang Technological University, School of Biological Sciences, Singapore,

(http://www.sbs.ntu.edu.sg/Pages/Home.aspx) has played a pivotal role in characterizing

dengue protein structures useful for drug design in collaboration with University of Marseille

and Novartis-NITD, and follows a close collaboration with Novartis-NITD in many aspects of

anti-dengue drug design. Specific research team: School of Biological Sciences, Dr. Julien

Lescar, http://www.sbs.ntu.edu.sg/AboutSBS/Faculty/Julien/Pages/Home.aspx

• National Chemical Laboratory (http://www.ncl-india.org/)and National Institute of

Virology (http://www.niv.co.in/) Pune, India, are developing together anti-dengue drugs from

Indian natural products using innovative and semi-automated fractionation procedures.

Specific research teams : Dengue Group, Drs. Dayaraj C. & Dr. P.S. Shah,

http://www.niv.co.in/annual_reports/Annual_Report_08-09/4%20chp%20Dengue

%20group.pdf

• Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh

City, Viet Nam. (http://www.tropicalmedicine.ox.ac.uk/viet-nam) This institution has been and

is conducting clinical trial of anti-dengue compounds, chloroquine and nucleoside analogue

(see Roche), respectively. It is currently the first and only center known in the world to

perform clinical trials on dengue drugs. Specific research team :

Dr. Cameron P. Simmons, http://www.tropicalmedicine.ox.ac.uk/cameron-simmons-2

Dr. Jeremy Farrar, http://www.tropicalmedicine.ox.ac.uk/jeremy-farrar

• Purdue University, Department of Medicinal Chemistry and Molecular Pharmacology,

School of Pharmacy and Pharmaceutical Sciences and the Purdue Cancer Center, West

Lafayette, Indiana, USA, (http://www.mcmp.purdue.edu/) in addition to its pioneering work

on dengue structural proteins, has performed virtual screening of compounds on the E protein

and identified thiazole compounds active in dengue infected cells. Specific research team :

Department of Medicinal Chemistry and Molecular Pharmacology, Dr. Carol B. Post

http://www.mcmp.purdue.edu/faculty/?uid=cbp

Department of Biological Sciences, Dr. Richard J. Kuhn,

http://www.bio.purdue.edu/people/faculty/index.php?refID=16

Department of Biological Sciences, Dr. Michael. G. Rossmann,

http://www.bio.purdue.edu/people/faculty/index.php?refID=81

27

Page 28: dengue

• Rega Institute Katholieke Universiteit Leuven (http://www.kuleuven.be/rega/rei/). The

Laboratory for virology and Experimental Chemotherapy has a long record of anti-dengue and

anti-flavirus compound isolation and characterization from very diverse sources, using dengue

virus infected cells. This lab and the "Centre for Drug Design and Discovery" (CD3) at

K.U.Leuven have received in 2009 a total of 2.8 million euro from the British Wellcome Trust.

They will apply these funds to the search for possible new medication for the treatment of

infections related to the dengue virus. The virology lab has and is also involved in large scale

EU projects targeting dengue (VIZIER-FP6, SILVER FP7 projects, respectively). Specific

research team : Laboratory of Virology and Chemotherapy, Dr. Johan Neyts,

http://www.kuleuven.be/rega/cmt/JN/group%20JN.html

• Shanghai Institute of Materia Medica, Shanghai, China (http://english.simm.cas.cn), The

Shanghai Institute of Materia Medica has reported that WSS45, a sulfated alpha-D-glucan

isolated from the medicinal plant Gastrodia elata, strongly interferes with Dengue 2 virus

infection in vitro, probably through the inhibition of virus binding to the host cell.Specific

research team : Laboratory of Immunopharmacology and Shanghai Institute of Materia

Medica, Dr. Zuo J.P.

• Southern Research Institute, Birmingham, AL, USA (http://www.southernresearch.org)

The Southern research Institute is a diversified network of collaborative centers for scientific

discovery and technology development. Their current capabilities implementation includes In

vitro services for studies related to dengue and West Nile viruses, amongst others.Specific

research team : Infectious Disease Program, Dr. James W. Noah,

http://www.southernresearch.org/life-sciences/biochemistry-molecular-biology/infectious-

disease-program

• State Key Laboratory of Virology, Wuhan Institute of Virology, China

(http://english.whiov.cas.cn/rh/rd/200907/t20090724_25180.html). The institute has identified

palmatin as a compound targeting the NS2b/NS3 protease, and has worked in collaboration

with the Wadsworth Center in the discovery and evaluation of anti-dengue compounds (see

Wadsworth Center). Specific research team : Dr. Zhiming Yuan

• Universidad de Buenos Aires, Departamento de Química Orgánica, Buenos Aires,

Argentina (http://www.qo.fcen.uba.ar) has a long record of isolation anti-dengue molecules

28

Page 29: dengue

from natural sources, as well as synthesis/hemi-synthesis of antivirals against dengue and

other hemorrhagic fever viruses. Specific research team: Dr. Elsa Damonte

• Université de la Mediterannée – Aix Marseille II - Centre National de la Recherche

Scientifique, Marseille, France (http://www.afmb.univ-mrs.fr) has a record of studies on

dengue replication and capping enzymes and their use in drug design. The virology

department is operating a screening platform with dengue as a top project including enzyme-,

and replicon-based assays, with a collaborative hit validation on dengue virus infected cells.

The virology department is involved in the EU-FP7 SILVER project having dengue drug

design as a major focus. Specific research team: Dr. Bruno Canard, http://www.afmb.univ-

mrs.fr/-Replicases-Virales-Structure-

• University of Berkeley, (http://sph.berkeley.edu/index.php). The Harris Laboratory, Division

of Infectious Diseases and Vaccinology, School of Public Health, University of California,

Berkeley (http://sph.berkeley.edu/faculty/harris.php) has a long-standing interest and track

record in many aspects of dengue research connected to antiviral research, such as the

evaluation of mycophenolic acid, interferons, and therapeutic monoclonal antibodies, and

definition of RNA elements in the 5' and 3' untranslated regions that are critical for translation

and replication. The Harris laboratory has established a mouse model of dengue pathogenesis

and antibody-dependent enhancement (ADE) of disease to study the potential for various

therapeutic candidates that 1) target the virus or host processes necessary for the viral life-

cycle, 2) interfere with the virus-antibody-Fc receptor interaction thereby inhibiting ADE, or

3) reduce dengue disease mediated by inflammation/vascular leakage. More recently, a

partnership with Nanoviricides Inc. has been set up to evaluate drugs in dengue virus-infected

cells and the mouse model of disease, the latter constituting a strong and active field of

research. Specific research team: Dr. Eva Harris, http://sph.berkeley.edu/faculty/harris.php

• University of Bristol, School of Medical Sciences (http://www.bris.ac.uk/fmvs/), has

developped replicons and infectious clones useful for mutation evaluation and drug sensitivity

studies. Specific research team : Department of Cellular and Molecular Medicine, Dr.

Andrew Davidson, http://www.bris.ac.uk/cellmolmed/staff/davidson.html

• University of Malaya, Department of Molecular Medicine, Faculty of Medicine, Malaysia

(http://umfacts.um.edu.my) has discovered and evaluated antiviral actions of flavanoid-

derived compounds on dengue virus type-2. Specific research team : Department of

29

Page 30: dengue

Molecular Medicine, Dr. Rohana Binti Yusof, http://umexpert.um.edu.my/papar_cv.php?

id=AAAJxnAAQAAAF9nAAO

• University of Texas Medical Branch, Department of Biochemistry and Molecular Biology,

Galveston, TX, USA, (http://www.utmb.edu/) The University of Texas Medical Branch,

Department of Biochemistry and Molecular Biology has launched the program "discovering

dengue drugs together" (http://www.utmb.edu/discoveringdenguedrugs-together/) and has

focused so far on antiviral discovery and design targeting the dengue protease. Specific

research team : Dr. Stanley J Watowich http://www.utmb.edu/scvd/staff.asp?ID=45

• Wadsworth Center, New York State Department of Health, and Department of Biomedical

Sciences, University at Albany, State University of New York, Albany, New York, USA

(http://www.wadsworth.org). The center and laboratory has been characterizing dengue

replicons, proteins and enzymes as targets for anti-dengue therapy, as well as discovering and

evaluating compounds, such as Brequinar, triaryl pyroazoline, PMO compounds (in

collaboration with Avi Biopharma), cyclosporin, a series of adenosine analogues related to

NITD008. Specific research team : Dr. Pei-Yong Shi. & Hongmin Li.

http://www.wadsworth.org/bms/index.html

• Washington University School of Medicine, Department of Medicine, St. Louis, USA,

(http://medschool.wustl.edu) has a long record in studies of dengue antibodies having

therapeutic potential. Specific research team : Diamond Lab Research, Department of

Molecular Microbiology, http://microbiology.wustl.edu/Research/diamondRES.html

30

Page 31: dengue

Part 4. The current industrial network of AV discovery

The first observation is that major pharmaceutical companies have not openly entered the field of

antivirals against dengue. No such company has any drug neither on their available product, nor on

their accessible list of products under development. However, some of these companies are developing

anti-dengue drugs, under two circumstances: either anti-dengue drugs were initially intended anti-

HCV drugs, or they are undertaking exploratory research and dengue field surveillance without

significant advertisement.

Pharmaceutical companies and R&D centers involved in dengue therapeutics

• Alios BioPharma (http://www.aliosbiopharma.com/) Alios BioPharma is developing novel

medicines to treat diseases in virology by activating pathways in the innate immune system.

Alios is pursuing the development of small-molecule and protein therapeutics. Small

molecules are of two general kinds: phosphate-protected nucleotides to ensure best

bioavailability through improved membrane crossing, and small RNaseL activators. The

company also develops modified interferons of improved properties, and although not directly,

dengue therapeutics is significantly addressed by Alios BioPharma activities and products.

• Alnylam Pharmaceuticals (http://w.alnylam.com) Alnylam is involved in the development of

RNAi-based innovative approaches targeting many different diseases. Although dengue is not

specifically mentionned, one of their focus is to obtain broad spectrum RNAi anti-viral

therapeutics against hemorrhagic fever viruses through collaborative biodefence programs.

• Apath (http://www.apath.com/) Apath is mainly a technology licensing company with a focus

on hepatitis C virus (HCV) and other medically important RNA viruses including, influenza

virus, respiratory syncytial virus (RSV), and hemorrhagic fever viruses. Apath contacts and

follows clients in the discovery and development of novel therapeutic products for the

treatment of viral infections, amongst which Dengue, West Nile and Yellow fever viruses are

mentioned. Apath has a proprietary antiviral screening platform, and all replicon technologies

useful for drug discovery studies.

• Arrow Therapeutics (http://www.astrazeneca.co.uk/) Arrow Therapeutics, Ltd., a

pharmaceutical company, focuses on the discovery and development of antiviral therapies. It

specializes in small molecule drugs with novel mechanisms of action, for respiratory syncytial

and hepatitis C virus, and has expressed interest and commitment in dengue antiviral research.

31

Page 32: dengue

• Avi BioPharma (http://www.avibio.com/) Avi BioPharma is developping modified

oligonucleotides of the phosphorodiamidate morpholino oligomers, conjugated or not to

peptides. These chemicals target RNA structures with great specificity, since some of their

best compounds are devoid of activity on West-Nile virus. The main focus of the company in

antiviral research is Influenza and Dengue.

• Biota (http://www.biota.com.au) Biota is an antiviral drug discovery and development

company focusing mainly on viral respiratory diseases (RSV, Rhinoviruses, and Influenza).

Although not directly involved in dengue antiviral research, Biota has discovered and is

developing a novel class of antiviral nucleoside drugs which inhibit the HCV polymerase.

• Biotron (http://www.biotron.com.au/) Biotron is focusing on "viroporins”, which are ion

channels involved in the ion traffic across membranes. Since virus replication is tighly

associated with membranes, small molecules blocking the ion channel activity of viroporins

are able to inhibit viral budding and replication. In the case of Dengue, the company is

focusing on the M protein.

• Botanic century (http://www.botaniccentury.com/index.asp) Botanic century and Phynova are

associated to investigate a plant extract (PYN-18) that has shown potent anti-dengue (as well

as anti-HCV) activity. PYN18 does not target viral absorption, replication, formation or

secretion of progeny virus. PYN18 appears to interfere with viral maturation such that

progeny virus is not infective to host cells.

• Canopus BioPharma (http://www.canopusbiopharma.com/) Canopus Biopharma is

developping CB5300 as a lead compound against Dengue. CB5300 is a product identified

long ago, used in the food industry since 1813, and generally recognized as safe (GRAS). It

was initially developped by Canopus BioPharma against HCV, and is thus a by product raising

interest in the field of antivirals against dengue. Although no information is available yet on

the nature of the product, it is expected to have a cellular target and systemic effect conferring

anti-dengue properties.

• Chimerix (http://www.chimerix-inc.com) Chimerix is an antiviral company screening

compounds to address several clinically significant viruses. Dengue virus is listed in their

currently active research program.

32

Page 33: dengue

• Center for Genetic Engineering and Biotechnology, CIGB (http://www.cigb.edu.cu/)

Although most of the efforts of the CIGB are oriented towards vaccine research, the CIGB is

developping antiviral molecules targeting dengue endocytic receptors, as well as peptides

targeting the NS3/NS2b protease. The CIGB is the first and only example of an institution

having a corporate activity (mixed with academic research) on dengue and other viral diseases

in low income countries.

• Functional Genetics (http://www.functional-genetics.com/) Functional Genetics is

developing antivirals (monoclonal antibodis and small molecules) that can treat or prevent a

broad spectrum of different viral diseases. Although not mentionned directly, dengue may

become a target of this company addressing mainly host cell targets.

• Genelabs technologies (http://www.genelabs.com) Genelabs technologies is an antiviral drug

discovery and design company focusing on HCV, and having advanced HCV nucleoside

inhibitors targeting the NS5b polymerase. Although not specifically mentionned, these

inhibitors may prove interesing for dengue. Genelabs has collaborated with Novartis and

Gilead in several scientific programs.

• Genodysse (http://www.genodyssee.com/), Genodyssee is a drug discovery company

specialized in the identification of host cell factors involved in response to diseases, and in

particular, viral infections. The company has isolated highly efficient interferons that will

probably raise interest beyond the initial intented use for HCV infections.

• International Center for Genetic Engineering and Biotechnology (ICGEB)

(http://www.icgeb.org/home.html) Beside their important reserach activity on Dengue vaccine

and diagnostics, the ICGEB has patented the use of activity Cissampelos pariera extracts

against Dengue, in a joint collaboration and application with Ranbaxy Research Laboratory,

now Daiichi Sankyo India Pharma Private Limited.

• Idenix Pharmaceuticals (http://www.idenix.com/) Idenix has no research and products

against dengue per se, but HCV compounds may find an additional or second life in dengue

antiviral research. This is particularly true for nucleoside analogues inhibitors of the HCV

NS5b RNA polymerase, which are very likely to inhibit the related dengue virus NS5 RNA

polymerase. In september 2010, the FDA had verbally informed Idenix that it should halt

study of two of those HCV compounds (IDX184 (completed Phase IIa) and IDX320

(completed Phase I)) due to three adverse events that had cropped up during a combination

33

Page 34: dengue

study of the two drugs. In the case of dengue, the short duration of the treatment may

accommodate these adverse effects, even more if the drugs do not show adverse effects when

give alone. In any case, re-evaluation of Idenix nucleoside analogue library may well uncover

interesting anti-dengue molecules. Novartis has 47% share in Idenix.

• Kineta (http://www.kinetabio.com/) Kineta is developing drug candidates that activate RIG-I,

a natural disease-fighting mechanism within the immune system. Research is focused on the

development of broad based antiviral products targeting, amongst others, hepatitic C and West

Nile viruses. Kineta has a proprietary screening platform

• Macrogenics (http://ww.macrogenics.com) Macrogenics discovers, develops and

commercializes antibody-based therapeutics spanning multiple therapeutic areas, including

immunology, oncology, respiratory, cardiometabolic and infectious diseases. A West Nile Mab

is under clinical phase II. For dengue, Macrogenics is developing an antibody for post-

exposure prophylaxis through funding by NIH.

• Nanoviricides (http://www.nanoviricides.com) The aim of Nanoviricides is to discover

compounds acting like microbicides, i.e., attacking enveloped virus particles and dismantling

them. Nanoviricides has established collaboration with USAMRIID to work on a large panel

of pathogenic human viruses, including dengue and flaviviruses. They have Identified drug

candidates showing efficacy in animals. In collaboration with Dr. E. Harris (University of

California, Berkeley), data has been obtained showing increased survival and decreased viral

load in a dengue virus-infected mouse model.

• Novartis and Novartis Institute of Tropical Diseases (http://www.novartis.com) Novartis is

one of the world largest drug discovery and design company. Novartis interest in Dengue is

carried mainly through the Novartis Institute of Tropical Diseases (NITD), which is mainly a

small-molecule drug discovery research institute dedicated to new treatments and prevention

methods for dengue, tuberculosis and malaria, and potentially by Idenix (into which Novartis

has ~45 % share). The goal of NITD is to make treatments readily available, without profit, to

patients of developing countries. NITD is by far the most active private institution. NITD has

published the greatest number (>50 in december 2010) of scientific publications and patents in

the dengue antiviral field since 2005. NITD is also the earliest institute having explicitly and

publicly put dengue as a priority. Initial efforts of NITD have focused on the dengue

NS2b/NS3 protease. NITD has conducted a large-scale HTS (1.4 million compounds and

identified interesting hits, but so far none has entered the development pipeline yet. For an

34

Page 35: dengue

unknown number of research teams, the impact on NS2b/NS3 inhibitor discovery of the

fabricated and erroneous crystal structure of dengue NS3 protease domain (published in

1999) remains to be evaluated. NITD has followed as many targets as possible from the virus

and also from the host, with significant impact in the field. These targets are the envelope

protein E (entry and fusion inhibitors), NS3 protease and helicase, NS4b association with

NS3, NS5 methyltransferase and RNA polymerase (for which 1 million compounds have been

screened), as well as conducted many studies (genome wide-, immunological-, membrane

studies, assays systems,...) that could identify or lead to cellular targets. Monoclonal

antibodies have been described with a potential therapeutic use.

NITD’s most advanced target and corresponding compounds is the NS5 MTase/polymerase.

NITD008 is an adenosine analogue first of a series studies by NITD, showing very interesting

properties, but abandoned because of unfavorable toxicity. A follow-up of this compound was

pursued with NITD449, further bonified with a prodrug approach to yield NITD203.

Unfortunately, the toxicity of the product neither reached an acceptable level. Follow-up of

this compound was pursued with NITD449, further bonified with a prodrug approach to yield

NITD203. The No-Observable-Adverse-Effect (NOAE) level was not overcome yet, though.

• Pharmasset Inc (http://www.pharmasset.com/) Pharmasset is an antiviral drug discovery,

design, and development company focusing mainly on HIV and HCV, but has several

nucleoside analogues (amongst which one is in collaboration with Roche) in clinical trails

against HCV that might eventually prove active against Flaviviridae at large, ie., dengue.

• PharmBridge (http://www.pharmabridgegroup.com/) See Drexel University for project and

compounds. Pharmabridge Inc. provides contract research and development activities for

pharmaceutical, biotechnology and agricultural industries.

• Prosetta Bioconformatics (http://www.prosetta.com) Prosetta Bioconformatics is a

biotechnology company exploring the potential of cell-free protein synthesizing systems for

drug discovery. Major focus is on antiviral research, and dengue targets have been studied (eg.

capsid formation) and followed up to find series of active compounds in dengue infected cells,

in collaboration with USAMRIID

• Replizyme Ltd (http://www.replizyme.com) Replizyme is a service company in the field of

antiviral research, and more specifically on viral polymerases. Both dengue and HCV purified

RNA polymerases are available as reagents and assays, as well as expertise to discover hits

and follow-up the hit-to-lead process.

35

Page 36: dengue

• Retrovirox (http://www.retrovirox.com) RetroVirox is a biotechnology company dedicated to

discovering new treatments for patients with infectious disease. The company focuses on

challenging human pathogens, dengue amongst other viruses. Antiviral assays against Dengue

virus are performed to determine the inhibitory activity of small-molecule compounds and

neutralizing antibodies for vaccine design.

• Roche (http://www.roche.com) Roche has entered very recently the dengue antiviral field with

a drug, Balapiravir, initially designed for HCV infections. It is a cytosine nucleoside analogue

dengue RNA-polymerase inhibitor, which has reached phase IIb against HCV, and been

abandoned because of haematological toxicity. As mentioned previously, the toxicity profile

may be irrelevant in the case of acute dengue infections, and thus, the compound has entered a

clinical trial in march 2010 entitled “Randomized, Double-Blind, Placebo-Controlled Study to

Evaluate the Safety and Efficacy of the Dengue Virus Polymerase Inhibitor (Balapiravir) in

Male Patients With Confirmed Dengue Virus Infection”. The trial is conducted by the Oxford

University Clinical Research Unit in Vietnam, the same institution which has evaluated

chloroquine in a clinical trial.

Likewise, Roche has been working on beta-d-2'-ethynyl-7-deaza-adenosine, a nucleoside

analogue presenting very potent inhibition properties of the Dengue virus RNA polymerase.

Studies were made using the active 5’-triphosphate of the drug in conjunction with purified

polymerases of the four dengue virus serotypes. This drug turned out to be the same as

NITD008, described in a recent paper, and abandoned because of unfavorable toxicity. It does

say, however, that a potential exists for nucleoside analogues targeting this essential dengue

enzyme.

• Sentinext therapeutics (http://www.sentinext.com)

Sentinext therapeutics is developing vaccines and immuno-therapeutics against dengue and

other viral diseases.

• SIGA technologies (http://www.siga.com) SIGA is anti-infectious company focusing mainly

on biodefence-concerning viruses. SIGA uses high-throughput screening technologies to

identify the most likely candidates for therapeutic intervention against, amongst others,

dengue virus. Small molecule inhibitors have been isolated and shown proof-of-concept

efficacy in a murine model.

36

Page 37: dengue

• Spring bank Pharmaceuticals (www.springbankpharm.com) Sping Bank Pharmaceuticals is

following a similar approach as Kineta, using nucleoside analogues turning on the RIG-I

innate immunity defense system against a broad range of viruses.

• Tamir (http://www.tamirbio.com/) Tamir is developing recombinant RNAses targeting cancer

cells as well as virus infected cells. RNA viruses and dengue viruses are amongst their

therapeutic targets. AC 03-636 has entered pre-clinical trials for gliomas and is under

investigation as a potentially broad spectrum antiviral.

• Tibotec (http://www.tibotec.com/) Tibotec is antiviral company, subsidiary of Johnson &

Johnson, known in the anti-HIV and anti-HCV field. Tibotec has shown interest and

commitment in dengue antiviral research.

37

Page 38: dengue

Part 5. Mapping the dengue drug design effort and needs

The location of the research entities described above in Part 3 and 4 are depicted within two

interactive maps at http://denguedrugs.com

The nature and location of academic dengue research centers

Examination of Map1 and Map 2 shows in a striking manner that the vast majority of dengue research

centers is NOT located in dengue-afflicted countries, neither at the academic level (Map 1) nor at the

corporate level (Map 2). The blatant and almost unique exception is NITD in Singapore.

Two continents heavily afflicted by dengue are absent from dengue antiviral research, namely Central

and South america (Latin America), and Africa.

Following the above discussion on natural products and biodiversity, if one considers that the tropical

countries containing the major share of natural molecules bearing anti-dengue potential, we note that

DENV geographical distribution superimposes very well with this natural biodiversity that should

provide drugs in the future.

Strikingly, nearly all low income country located between the tropics has a department of natural

products in its university. This is, in part, due to the natural abundance of plants, the

ethnopharmacological practices, and the impact of the Convention on Biological Diversity (december

1993), establishing access and benefit sharing guidelines. For example, Africa has a well organized

network named Napreca consisting of universities and research organisms dedicated to natural product

research. Napreca (see http://napreca.net/) involves in east and central Africa Bostwana, Cameroon,

Congo, Democratic Republic of Congo, Ethiopia, Kenya, Madagascar, Rwanda, Tanzania, Uganda,

and Zimbabwe.

It is thus obvious that in the future, dengue research will be also conducted in situ as more bottlenecks

(cost, education, technology, policy) are overcome.

Patent activity of the last 6 years (2005-2010)

The list of patents directly relevant to drug design, having dengue as a direct or indirect focus (ie., as a

by product of HCV, or a general antiviral method, …) is reported in Annex 1.

An increase in the number of patent is detectable in the last two years, with a number averaging 20+

patents, whereas this number by about half the years before. Another interesting information is the

nature and content of the patents is evolving.

38

Page 39: dengue

Indeed, one can see in Figure that the proportion of patents having “dengue” or “Flavivirus” (or

“Flaviviridae” when it is specified that it is in addition to HCV) is strongly increasing over the years,

going from 13 % in 2005 to more than 50 % in 2009 and 2009. If this trend continues, on can safely

say that something is happening in the dengue drug design field. Dengue is becoming a major focus,

not a by-product of antiviral research on other clinically and economically important viruses.

Figure 2. The past 6-years patent activity

related antivirals against viruses

amongst which dengue, and patents having

“dengue” or “flavivirus” in their title.

Trends, strengths, and weaknesses of the anti-dengue drug research/development field

The field of dengue drug research has emerged in the past 5 years and is probably here to stay. The

knowledge accumulated both in the drug design field and on DENV specifically allows efficient drugs

to be discovered. It is clear that although perfectible, the scientific and drug design community

possesses tools to synthesize, select, improve, and evaluate drugs in the laboratory. What is the nature

of the bottlenecks, where are they, or more exactly, where have they moved to?

The once often mentioned patient cohort availability for clinical trials has been recently overcome.

The market issue seems to be less significant also, as having private companies investigating the field

is the best and safest indicator of a sizeable market.

On the scientific point-of-view, perhaps the most important bottleneck is that of appropriate animal

model. These are of two kinds, i) a model for infection and protective action of a given drug, ii) a

disease model, that could, as discussed above, also yield interesting therapeutic avenues for tailored

response modifier drugs.

Mice model exist (eg. AG129 mouse), but they have their limitations (eg., low and short viremia).

Taking advantage of the possibility to manipulate mice genetically, other models will have to be

39

Page 40: dengue

developed. Likewise, monkey models exist and would benefit to be optimized regarding which viral

strains (epidemic, sylvatic, clinical, reference, etc…) are they best for.

One last potential limitation to drug development is that dengue disease is perhaps majoritarily a

pediatric disease, and most of the avenues taken in drug design have involved adult patients. This is

not a simple drug-dosage issue. For example, no genome wide research has been conducted comparing

infected infants versus adults. The whole dengue drug research must be at all times evaluated with this

issue in mind, as a potential specific toxicity or adverse effect in children would devastate the still

fragile antiviral drug design field.

Beyond dengue : other relevant viral infections

After having served as an initial model for HCV, dengue research will certainly benefit to research on

other viruses. In fact, dengue provides to the drug designer significant difficulties that may not be

found for other virus infections (eg., pediatric disease, short viremia, disseminated target organs,

encephalitic forms,…). In that respect, all the great effort made for dengue will disseminate to specific

fields, and in first instance, for flaviviruses. In the case of Yellow fever virus, for which there is a very

efficient vaccine, it is not known if the use of a potent antiviral immediately after the onset of

symptoms would save lives from this devastating illness. For encephalitic viruses (JEV, TBE, WNV,

…), the question remains the same regarding neurological symptoms and disease severity.

In the most conserved flavivirus protein (NS5), the amino acid divergence between dengue serotypes 1

- 4 is 20- to- 26% whereas West Nile virus NS5 has only ~33% amino acid divergence with that of

dengue (Table 1)

Table 1. Amino acid conservation between

dengue and West-nile (kunjin isolate) for

the whole ORF (upper part in black) and

NS5 (red and green).

It is thus very likely that drugs targeting conserved flaviviral proteins or generally conserved virus –

host interaction pathways will have a broad activity spectrum amongst flaviviruses. Although this

observation holds for identified BSL-4 viruses, it is important to note that a very significant number of

encephalitis of unknown etiology occur in the tropics. Some are of viral (and flaviviral) origin, and

may give rise to a new emerging flavivirus.

Last, beyond dengue are other viruses that add to the dengue burden. There are no large scale studies

of dengue co-infections with other viral or bacterial/fungal/parasitic pathogenic agents. What about

dengue and HIV? dengue and measles? dengue and malaria? dengue and other seemingly mild

40

Page 41: dengue

pediatric diseases? Etc…We need clearer pictures of these co-infections, as the drug-drug interaction

field is also quite young. This adds complexity to issues related to the patient’s background and its

effect on dengue disease outcome.

How to promote the development of dengue antiviral research?

As shown in the list of pharmaceutical companies, most of the latter are small-size, risk taking

companies investigating research field that are not covered by large pharmaceutical companies. They

are created upon discovery of an active compound which is then patented, or they are investigating

novel avenue of research uncovered by academic discoveries at large (eg., host factors, innate

immunity, etc…). In the former case, they take a “traditional” development scheme, which is

notoriously long, costly, and risky. This may prove even harder in the case of dengue than HCV or

other viruses, and not surprisingly, these companies are not so small… In the latter case (ie.,

innovative novel avenues), they may either subcontract discoveries to larger companies, or grow to a

larger size. There is also a high risk associated to this approach, as business development example do

not abound in these novel fields, and they may anyway meet with a traditional drug design scheme

later on.

Therefore, one can see two ways of favoring the emergence of dengue drug design companies: The

first is a strong academic support to innovative and novel approaches presenting space yet unexplored

by large pharmaceutical companies. This are mainly pathogenesis, genome-wide, host-response, and

innate immunity studies. Significant discoveries will undoubtedly generate new start-ups from

scientists and corporate entrepreneurs. The second incentive is to make chemical molecules available

to whoever has capacity, will, and structure to discover drugs. Natural products are unbeatable in many

aspects described above, and through existing university networks in dengue afflicted countries, they

have the capacity to federate research effort, and foster scientific, medical, and economical

development. As Asia has already made significant efforts in this direction, capacity building remains

to be established in Africa and Latin America.

41

Page 42: dengue

Annex 1. References

Recommended readings

• Dengue (2008) Tropical Medicine: Science and Practice, Vol. 5, Scott B. Halstead Editor,

Imperial College Press.

• Gubler, D., Kuno, G., and Markoff, L. (2007) Flaviviruses, in Fields Virology, Vol.1 Fifth

edition, LWW, Knipe DM and Howley PM, Eds.

• Noble, C. G., Chen, Y. L., Dong, H., Gu, F., Lim, S. P., Schul, W., Wang, Q. Y., Shi, P. Y.

(2010), Stategies for the development of DENV inhibitors, Antiviral Research, 85, 450-462.

Additional references cited in the text :

1. Eggink, D., B. Berkhout, and R. W. Sanders. Inhibition of HIV-1 by Fusion Inhibitors. Curr

Pharm Des 16:3716-28.

2. Elion, G. B., P. A. Furman, J. A. Fyfe, P. de Miranda, L. Beauchamp, and H. J. Schaeffer.

1977. Selectivity of action of an antiherpetic agent, 9-(2-hydroxyethoxymethyl) guanine. Proc

Natl Acad Sci U S A 74:5716-20.

3. Kingston, D. 2011. Modern Natural Products Drug Discovery and IT Relevance to

Biodiversity Conservation. J. Nat. Prod in the press.

4. Newman, D. J., and G. M. Cragg. 2007. Natural products as sources of new drugs over the

last 25 years. J Nat Prod 70:461-77.

5. Pevear, D. C., T. M. Tull, M. E. Seipel, and J. M. Groarke. 1999. Activity of pleconaril

against enteroviruses. Antimicrob Agents Chemother 43:2109-15.

6. Saklani, A., and S. K. Kutty. 2008. Plant-derived compounds in clinical trials. Drug Discov

Today 13:161-71.

7. Stittelaar, K. J., J. Neyts, L. Naesens, G. van Amerongen, R. F. van Lavieren, A. Holy, E.

De Clercq, H. G. Niesters, E. Fries, C. Maas, P. G. Mulder, B. A. van der Zeijst, and A. D.

Osterhaus. 2006. Antiviral treatment is more effective than smallpox vaccination upon lethal

monkeypox virus infection. Nature 439:745-8.

8. Taubenberger, J. K., A. H. Reid, A. E. Krafft, K. E. Bijwaard, and T. G. Fanning. 1997.

Initial genetic characterization of the 1918 "Spanish" influenza virus. Science 275:1793-6.

42

Page 43: dengue

Annex 2. Patents

The search was made with « dengue & antiviral », scientific publications in journals eliminated, and

patents kept and numbered for each year. Sometimes, the patent is applied or obtained in two different

flavors (according to, eg., countries for which it has been granted), but a single mention has been

reported here. Only patents related to dengue antiviral therapy (not diagnostics nor vaccine) are

mentioned. Sometime a method, compound or a target is relevant to dengue as a « side » virus, ie.,

intended at first against another virus.

2010 – 18 patents

1- Preparation of azaindole compounds and methods for antiviral treatment. Maccoss,

Malcolm; Njoroge, F. George; Nomeir, Amin; Chen, Guangming; Karp, Gary Mitchell; Lennox,

William Joseph; Li, Chunshi; Morrill, Christie; Paget, Steven D.; Ren, Hongyu; Zhang, Nanjing;

Zhang, Xiaoyan. (Schering Corporation, USA; PTC Therapeutics, Inc.). PCT Int. Appl. (2010),

228pp. CODEN: PIXXD2 WO 2010117935 A1 20101014 Designated States W: AE, AG, AL, AM,

AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK,

DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE,

KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW,

MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK,

SL, SM, ST, SV, SY, TH, TJ. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR,

IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, SM, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN,

TD, TG. Patent written in English. Application: WO 2010-US29928 20100405. Priority: US

2009-166883 20090406. CAN 153:505795 AN 2010:1277282 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Abstract

The invention is directed to compds. of formula I and forms and pharmaceutical compns. thereof

useful for treating a viral infection, or for affecting viral activity by modulating viral replication.

Compds. of formula I wherein W1, W2, W3 and W4 are independently N, CR1 and NO, provided that

one to three of W1 - W4 are N and the remainder are CR1; X is H, halo, CN, NO2, etc.; Y is

(un)substituted aryl, (un)substituted heteroaryl, and (un)substituted heteroaryl-1-oxide; Z is C1-8

alkyl, C2-8 alkenyl-C1-8 alkyl, C2-6 alkynyl-C1-8 alkyl, etc.; R1 is H, halo, OH, CN, NO2, etc.; and

free acids, free bases, salts, hydrates, solvates, clathrates, isotopologues, racemates, enantiomers,

diastereoisomers, stereoisomers and polymorphs thereof, are claimed. Example compd. II was prepd.

43

Page 44: dengue

by a general procedure (procedure given). All the invention compds. were evaluated for their antiviral

activity. From the assay, it was detd. that compd. II exhibited an IC50 value of less than about 0.5 μM.

2- Polynucleotides comprising codon-optimized prME genes from dengue virus types 1-4, their

sequences and use in generation of viral-like particles for drug screening, immunization or

disease treatment. Wang, Peigang; Altmeyer, Ralf Marius; Nal-Rogier, Beatrice Therese Marie;

Kudelko, Mateusz; Despres, Philippe. (Institut Pasteur, Fr.; HKU-Pasteur Research Centre). Can.

Pat. Appl. (2010), 43pp. CODEN: CPXXEB CA 2658259 A1 20100912 Patent written in

English. Application: CA 2009-2658259 20090312. Priority: . CAN 153:424495 AN

2010:1193018 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention provides polynucleotides comprising codon-optimized premembrane glycoprotein M

and E glycoprotein genes (prME) from dengue virus types 1-4 for enhanced expression in mammalian

cells, and their use in generation of virus-like particles (VLPs). The invention also provides for the

use of said VLPs in: (a) eliciting a specific anti-dengue immune response against dengue-assocd.

diseases or infections; (b) screening for agents that inhibit dengue virus prodn.; and (c) treating or

preventing a dengue-assocd. diseases. The invention further provides the sequences of said

polynucleotides that comprise the codon-optimized prME genes. The examples showed that the

optimized DV1 prME gene in transformed HeLa cells enhanced the expression of prME glycoproteins

and thereof facilitated the generation of DV VLPs with native viral proteins

3- Thienopyridine derivatives for the treatment and prevention of dengue virus infections.

Byrd, Chelsea M.; Dai, Dongcheng; Jordan, Robert; Hruby, Dennis E. (Siga Technologies, Inc.,

USA). PCT Int. Appl. (2010), 130pp. CODEN: PIXXD2 WO 2010099166 A1 20100902

Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA,

CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM,

GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU,

LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ. Designated States RW: AT, BE,

CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, SM, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2010-US25183 20100224. Priority: US 2009-156132 20090227. CAN 153:375216 AN

2010:1095820 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

44

Page 45: dengue

Abstract

Methods and pharmaceutical compns. for treating viral infections, by administering certain

thienopyridine deriv. compds. in therapeutically effective amts. are disclosed. Methods of using the

compds. and pharmaceutical compns. thereof are also disclosed. In particular, the treatment and

prophylaxis of viral infections such as caused by flavivirus is disclosed, i.e., including but not limited

to, Dengue virus, West Nile virus, yellow fever virus, Japanese encephalitis virus, and tick-borne

encephalitis virus.

4- Antibodies for diagnosis and treatment of flaviviral infections. Vasudevan, Subhash; Lescar,

Julien; Rajamanonmani, Ravikumar. (Nanyang Technological University, Singapore). PCT Int.

Appl. (2010), 107pp. CODEN: PIXXD2 WO 2010093335 A1 20100819 Designated States W:

AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR,

CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID,

IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG,

MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL, PT, RO, RS, RU, SC, SD,

SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI,

FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, SM, TR, BF, BJ, CF, CG, CI, CM, GA, ML,

MR, NE, SN, TD, TG. Patent written in English. Application: WO 2010-SG49 20100210.

Priority: US 2009-151301 20090210. CAN 153:331827 AN 2010:1039949 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The authors disclose the prepn. of an antibody raised against domain III of dengue virus envelope

glycoprotein (E). The antibody is capable of (1) neutralizing dengue virus; (2) binding to all dengue

serotypes, and (3) cross-reacting with West Nile virus.

5- Purine nucleoside monophosphate prodrugs for treatment of cancer and viral infections.

Cho, Jong Hyun; Coats, Steven J.; Schinazi, Raymond F.; Zhang, Hongwang; Zhou, Longhu. (RFS

Pharma, LLC, USA; Emory University). PCT Int. Appl. (2010), 143pp. CODEN: PIXXD2 WO

2010091386 A2 20100812 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG,

BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES,

FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA,

LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO,

NZ, OM, PE, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL,

NO, PT, SE, SM, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in

45

Page 46: dengue

English. Application: WO 2010-US23563 20100209. Priority: US 2009-150628 20090206. CAN

153:311457 AN 2010:1002272 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Purine nucleoside monophosphates I, wherein R1 is an atom or a group removed in vivo to form OH

when administered as the parent nucleoside, for example halogen, OR, NR2, SR, OCOR, CHCOR,

N(COR)COR, SCOR, OCOOR, NHCOORR is H, alkyl, haloalkyl, alkoxy, alkenyl, alkynyl,

cycloalkyl, aryl, heteroaryl, alkylaryl, arylalkyl; W is N, CH, CF, CCN, CC…CH, CC(O)NR2;Y is O,

S; Z is CL2, CL2CL2, CL2OCL2, CL2SCL2, CL2O, OCL2, CL2NHCL2; L is H, F, alkyl, alkenyl,

alkynyl; A is O, S, CH2, CHF, CF2, C=CH2, C=CHF, C=CF2; R2 and R3 are independently OR8; R8

is alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl; R5, R6, R4'-R7' are independently H, halogen, OH,

SH, NH2, NHOH, NHNH2, N3, COOH, CN, COMH2, CSNH2, ester, alkyl, R9, OR9, SR9, SSR9,

NHR9, NR92; R9 is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, arylalkyl; were prepd. and used

for treating or preventing cancer and viral infections. Thus, nucleotide II was prepd. and tested in

vitro as antitumor and antiviral agent. The title compds. show potent antiviral activity against HIV-1,

HIV-2, HCV, Norovirus, Saporovirus, HSV-1, HSV-2, Dengue virus, Yellow fever, and HBV.

6- Preparation of 1'-substituted carba-nucleoside analogs as antiviral agents carba-nucleoside

analogs for antiviral treatment. Butler, Thomas; Cho, Aesop; Kim, Choung U.; Xu, Jie. (Gilead

Sciences, Inc., USA). U.S. Pat. Appl. Publ. (2010), 91pp. CODEN: USXXCO US 2010203015

A1 20100812 Patent written in English. Application: US 2010-702957 20100209. Priority: US

2009-151248 20090210. CAN 153:287244 AN 2010:1001798 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Abstract

Provided are thieno[3,4-d]pyrimidin-7-yl and furo[3,4-d]pyrimidin-7-yl ribosides, riboside phosphates

and prodrugs thereof as well as intermediates and methods of prepn. The compds., compns., and

methods provided are useful for the treatment of Flaviviridae virus infections. 1'-Substituted carba-

nucleoside analogs I, wherein R1-R6 are independently H, OR, NR2, N3, CN, NO2, S(O)nR, halogen,

alkyl, carbocyclyl-alkyl, alkenyl, alkynyl, arylalkyl; any two of R1-R6 on adjacent carbo when taken

together are O(CO)O or when taken together with the ring carbon to which they are attached form a

double bond; n is 0-2; R is H, alkyl, alkenyl, alkynyl, arylalkyl, carbocyclyl-alkyl, acyl, carboxylate

ester, amide, thio ester, sulfonyl, sulfoxide, sulfonamide; R7 is H, acyl, carboxylate ester, amide,

sulfonyl, sulfonamide, substituted phosphorus; R8-R10 are independently halogen, substituted amine,

N3, NO, NO2, CHO, CN, substituted imine, substituted oxime, amide, sulfonamide, carboxylate ester,

x2 is S, O, S(O), SO2; were prepd. and used as antiviral agents. The compds., compns., and methods

provided are useful for the treatment of Flaviviridae virus infections, particularly hepatitis C

46

Page 47: dengue

infections. Thus, nucleoside II was prepd. and tested as antiviral agent (EC50 10-100 μM), wherein

the viral infection is caused by a virus selected from the group consisting of dengue virus, yellow

fever virus, West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus, Kunjin virus,

Murray Valley encephalitis virus, St. Louis encephalitis virus, Omsk hemorrhagic fever virus, bovine

viral diarrhea virus, Zika virus and Hepatitis C virus.

7- Anti-dengue activity of Cissampelos pareira extracts. Bhatnagar, Pradip Kumar; Katiyar,

Chandra Kant; Khanna, Navin; Upadhyay, Dilip Jatashankar; Swaminathan, Sathyamangalam;

Srinivas, Kona; Sharma, Navin; Kanaujia, Anil; Sood, Ruchi; Singhal, Smita; Shukla, Gyanesh;

Duggar, Rajeev; Pareek, Pawan Kumar; Singh, Yogendra; Khan, Seema; Raut, Rajendra. (Ranbaxy

Laboratories Limited, India; International Centre for Genetic Engineering and Biotechnology;

Department of Biotechnology). PCT Int. Appl. (2010), 26pp. CODEN: PIXXD2 WO

2010084477 A1 20100729 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG,

BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES,

FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA,

LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO,

NZ, OM, PE, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL,

NO, PT, SE, SM, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in

English. Application: WO 2010-IB50299 20100123. Priority: IN 2009-DE141 20090123. CAN

153:241902 AN 2010:943245 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention relates to the anti-dengue activity of the Cissampelos pareira exts.

Pharmaceutical compns. comprising exts. of C. pareira and processes for the prepn. of exts. are also

provided. Methanolic ext. of C. pareira showed antiviral activity against all the four serotypes of

dengue (DENV1, DENV2, DENV3 and DENV4) in conventional assay with PRNT50 values in the

range of 1.2-11.1 μg/mL.

8- Targeting and inhibiting replication of dengue virus type I using siRNA. Wu, Xinwei; Wang,

Ming; Du, Lin; Yue, Jinya; Jiang, Liyun. (Guangzhou Disease Prevention and Control Center, Peop.

Rep. China). Faming Zhuanli Shenqing (2010), 11pp. CODEN: CNXXEV CN 101781651 A

20100721 Patent written in Chinese. Application: CN 2009-10036705 20090116. Priority: . CAN

153:306789 AN 2010:926365 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

47

Page 48: dengue

Patent Family Information

Patent No. Kind Date Application No. Date

CN 101781651 A 20100721 CN 2009-10036705 20090116

Priority Application

CN 2009-10036705 20090116

Abstract

This invention relates to a method for targeting and inhibiting the replication of dengue virus type I

using siRNA. The invention provides the sequences of the siRNA mols. used for inhibiting the

replication of dengue virus. The siRNA sequence is screened from those that are designed according

to dengue virus type I genome, and can inhibit dengue virus replication, and prevent cells from being

damaged by dengue virus.

9- Inhibition of viral RNA interaction with host proteins in the inhibition of viral replication in

the treatment of infection. Brinton, Margo A.; Emara, Mohamed M.; Li, Wei. (Georgia State

University Research Foundation, USA). U.S. Pat. Appl. Publ. (2010), 41pp., Cont.-in-part of U.S.

Ser. No. 654,273. CODEN: USXXCO US 2010166704 A1 20100701 Patent written in English.

Application: US 2008-82095 20080407. Priority: US 2002-407105 20020830; US 2003-654273

20030902; US 2007-910611 20070406. CAN 153:135758 AN 2010:820082 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20100166704 A1 20100701 US 2008-82095 20080407

US 20040162252 A1 20040819 US 2003-654273 20030902

Priority Application

US 2002-407105P P 20020830

US 2003-654273 A2 20030902

US 2007-910611P P 20070406

Abstract

Methods of treating viral infection, specifically a flavivirus infection, by blocking the interaction of

viral RNA with host factors to prevent viral replication are described. Specifically, the interaction of

the host TIA-1 and TIAR antigens with the viral RNA 3'-stem-loop structure is identified as a target.

TIAR is shown to be the primary protein binding to the stem-loop structure. Knockout of the host

TIAR gene lowers the efficiency of viral replication in infected cells.

48

Page 49: dengue

10- Non-dividing cell-based assay for high throughput antiviral compound screening. Uprichard,

Susan L.; Yu, Xuemei; Sainz, Bruno, Jr. (University of Illinois, USA). U.S. Pat. Appl. Publ. (2010),

17pp. CODEN: USXXCO US 2010099079 A1 20100422 Patent written in English. Application:

US 2009-566074 20090924. Priority: US 2008-100540 20080926. CAN 152:493425 AN

2010:507665 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20100099079 A1 20100422 US 2009-566074 20090924

Priority Application

US 2008-100540P P 20080926

Abstract

The invention features a cell-based assay that recapitulates all aspects of a viral lifecycle for use in

identifying antiviral agents. The assay employs synchronized, non-dividing host cells and a

fluorescence resonance energy transfer peptide substrate for monitoring endogenous viral protease

activity, which is indicative of viral infection kinetics.

11- Dengue virus neutralizing antibodies. Lanzavecchia, Antonio. (Institute for Research In

Biomedicine, Switz.). PCT Int. Appl. (2010), 47pp. CODEN: PIXXD2 WO 2010043977 A2

20100422 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE,

GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS,

LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG,

PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, SM, TR,

BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application:

WO 2009-IB7372 20091013. Priority: US 2008-104911 20081013. CAN 152:499422 AN

2010:503823 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The authors disclose the prepn. and characterization of antibodies and antigen-binding fragments that

neutralize dengue virus infection without contributing to antibody-dependent enhancement of dengue

virus infection.

12- Preparation of nucleoside cyclic phosphates as antiviral agents. Du, Jinfa; Nagarathnam,

Dhanapalan; Pamulapati, Ganapati Reddy; Ross, Bruce S.; Sofia, Michael Joseph. (Pharmasset, Inc.,

USA). U.S. Pat. Appl. Publ. (2010), 73pp.; Chemical Indexing Equivalent to 152:57501 (WO).

49

Page 50: dengue

CODEN: USXXCO US 2010081628 A1 20100401 Patent written in English. Application: US

2009-479075 20090605. Priority: US 2008-60683 20080611; US 2008-140369 20081223; US

2008-140317 20081223. CAN 152:381685 AN 2010:406750 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Abstract

Cyclic phosphate of nucleoside derivs. I, were prepd., wherein R1 is H, alkyl, cycloalkyl, alkaryl, aryl,

halo-alkyl, halo-alkenyl, halo-alkynyl, alkoxy, halo-alkoxy, CO2H, carboxylate, CONH2, substituted

amide, CH=CHCO2H, CH=CH-carboxylate; R2 is H, alkyl, CN, Me, vinyl, O-alkyl, OMe, OEt,

hydroxy-alkyl, CHwF, N3, CH2CN, CH2N3, CH2NH2, CH2NHMe, CH2NMe2, ethynyl-alkyne,

halogen; R3 is H, Me, CH2F, CHF2, CF3, F, CN; X is H, OH, F, OMe, halogen, NH2, N3; B is

nucleobase; were prepd. and used for the treatment of viral infections in mammals, which is a compd.,

its stereoisomers, salts (acid or basic addn. salts), hydrates, solvates. Thus, nucleotide II was prepd.

and used as antiviral agent.

13- Novel imino sugar derivatives demonstrate potent antiviral activity and reduced toxicity.

Block, Timothy M.; Chang, Jinhong; Xu, Xiaodong. (Institute for Hepatitis and Virus Research,

USA). PCT Int. Appl. (2010), 29pp. CODEN: PIXXD2 WO 2010027996 A1 20100311

Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA,

CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM,

GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU,

LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM. Designated States RW: AT, BE,

CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, SM, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2009-US55658 20090902. Priority: US 2008-190618 20080902. CAN 152:327087 AN

2010:306482 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Imino sugars, such as deoxynojirimycin (DNJ), are glucose analogs that selectively inhibit cellular α-

glucosidase I and II (enzymes that process N-linked glycans in glycoprotein) and exhibit broad

spectrum antiviral activities against many enveloped viruses. To develop' imino sugars with more

potent antiviral activity as well as improved toxicity profile, OSL-9511 (N-pentyl(1-

hydroxycyclohexyl)-DNJ) was modified by diversifying the nitrogen linked alkylated side chain.

Furthermore, these new compds. were shown to be active against Dengue virus (DV) and West Nile

50

Page 51: dengue

virus (WNV) infection in BHK cells with potent anti-DV activity having submicromolar EC50 values

and SI of greater than 900. These compds. represent a new generation of imino sugars and their

analogs, having IS application in the clin. treatment of infection by DV and other members of

flaviviridae.

14- preparation of alkynyl nucleoside analogs for use as antivirals treating HCV and Dengue

viruses. Chen, Yen Liang; Duraiswamy, Jeyaraj; Haller, Sarah; Keim, Matthias; Kondreddi,

Ravinder Reddy; Yin, Zheng. (Novartis AG, Switz.). PCT Int. Appl. (2010), 121pp. CODEN:

PIXXD2 WO 2010015643 A1 20100211 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ,

BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC,

EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP,

KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA,

NG, NI, NO, NZ, OM, PE, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY,

TJ, TM. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC,

MT, NL, NO, PT, SE, SM, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent

written in English. Application: WO 2009-EP60125 20090804. Priority: EP 2008-161933

20080806. CAN 152:239231 AN 2010:179470 CAPLUS (Copyright (C) 2010 ACS on SciFinder

(R))

Abstract

Title compds. I, wherein X is CH or an optionally substituted alkyl, alkenyl, alkynyl, aryl, cycloalkyl,

heterocycloalkyl, heteroaryl, halogen, cyano, nitro, hydroxy, alkoxy, alkylthio, amino, alkylamino,

carboxy, carboxamide or alkyloxycarbonyl carbon atom; R1 is halogen, (un)substituted amine, or an

(un)substituted ether; R2 is H, halogen, or an (un)substituted amine; R3 is H, alkyl, alkenyl, alkynyl,

aryl, cycloalkyl, heterocycloalkyl or heteroaryl, each of which is optionally substituted with one or

more substituents; R4 is H, acyl or an amino acid ester; R5 is H, acyl or an amino acid ester are prepd.

as antiviral prodrugs. Thus, II was prepd. and demonstrated a dose response inhibition with a dosage

of 32mg/kg bid giving a redn. of viremia of 113 fold. Further, I can be successfully employed as

prodrugs treating Flaviviridae family viral infections selected from the group consisting of dengue

virus, yellow fever virus, West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus,

Kunjin virus, Murray Valley encephalitis, St Louis encephalitis, Omsk hemorrhagic fever virus, bovine

diarrhea virus, Zika virus and Hepatitis C virus. Finally, selected compds. were analyzed for their X-

ray powder diffraction patterns, IR spectrum, differential scanning calorimetry thermograms and

thermogravimetric anal. curve to enhance the most practical method of pharmaceutical drug delivery.

51

Page 52: dengue

15- Preparation of alkynyl nucleoside analogs for use as antivirals treating HCV and Dengue

viruses. Chen, Yen-Liang; Duraiswamy, Jeyaraj; Kondreddi, Ravinder Reddy; Yin, Zheng.

(Novartis AG, Switz.). PCT Int. Appl. (2010), 120pp. CODEN: PIXXD2 WO 2010015637 A1

20100211 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE,

GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS,

LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG,

PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, SM, TR,

BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application:

WO 2009-EP60114 20090804. Priority: EP 2008-161941 20080806. CAN 152:239230 AN

2010:179431 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Title compds. I, wherein X is CH or an optionally substituted alkyl, alkenyl, alkynyl, aryl, cycloalkyl,

heterocycloalkyl, heteroaryl, halogen, cyano, nitro, hydroxy, alkoxy, alkylthio, amino, alkylamino,

carboxy, carboxamide or alkyloxycarbonyl carbon atom; R1 is halogen, (un)substituted amine, or an

(un)substituted ether; R2 is H, halogen, or an (un)substituted amine; R3 is H, alkyl, alkenyl, alkynyl,

aryl, cycloalkyl, heterocycloalkyl or heteroaryl, each of which is optionally substituted with one or

more substituents; R4 is H or a halogen are prepd. as antiviral prodrugs. Thus, II was prepd. and

tested for its cell-based Flavivirus immunodetection assay, HCV replication assay, for activity against

dengue infection and a clin. trial protocol (no data). Further, I can be successfully employed as

prodrugs treating Flaviviridae family viral infections selected from the group consisting of dengue

virus, yellow fever virus, West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus,

Kunjin virus, Murray Valley encephalitis, St Louis encephalitis, Omsk hemorrhagic fever virus, bovine

diarrhea virus, Zika virus and Hepatitis C virus.

16- Method for preparing anti-dengue virus oral preparation from magnetic alginate sodium

drug loaded microspheres. Huang, Yunqing. (Peop. Rep. China). Faming Zhuanli Shenqing

Gongkai Shuomingshu (2010), 20pp. CODEN: CNXXEV CN 101632688 A 20100127 Patent

written in Chinese. Application: CN 2008-10134202 20080721. Priority: . CAN 152:271275 AN

2010:129001 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

CN 101632688 A 20100127 CN 2008-10134202 20080721

Priority Application

52

Page 53: dengue

CN 2008-10134202 20080721

Abstract

The method comprises prepg. water sol. Chinese medicine inclusion microsphere fines, prepg. alc. sol.

Chinese medicine inclusion microsphere fines, mixing two fines, processing to obtain capsules or

tablets. The water sol. Chinese medicine inclusion microsphere is prepd. by mixing β-cyclodextrin,

hypericin, Buffalo Horn ext. and Carapax Amydae ext. together in agitator under frequency of 50-200

r/min for 5-15 min, adding Fe3O4 powder, stirring for 30-90 min, setting the frequency to 50-500

r/min, stirring, adding water, stirring to obtain slurry, adding the slurry in supermol. envelope machine,

hydrating under frequency of 1000-3000 r/min for 30-120 min, drying at 40-65ϒ for 24 h and

pulverizing. The alc. sol. Chinese medicine inclusion microsphere is prepd. by mixing β-cyclodextrin

and artemisinin together in agitator under frequency of 50-200 r/min for 5-15 min, adding Fe3O4

powder, stirring for 30-90 min, setting the frequency to 50-500 r/min, stirring, adding 50-95% ethanol,

stirring to obtain slurry, adding the slurry in supermol. envelope machine, hydrating under frequency

of 1000-3000 r/min for 30-120 min, drying at 40-65ϒ for 24 h and pulverizing. The obtained anti-

dengue virus oral prepn. is used to treating sepsis by resisting dengue fever virus DNA replication and

RNA transcription.

17- Preparation of nucleoside phosphoramidate prodrugs as antiviral agents. Sofia, Michael

Joseph; Du, Jinfa; Wang, Peiyuan; Nagarathnam, Dhanapalan. (Pharmasset, Inc., USA). U.S. Pat.

Appl. Publ. (2010), 77pp.; Chemical Indexing Equivalent to 149:426212 (WO). CODEN:

USXXCO US 2010016251 A1 20100121 Patent written in English. Application: US 2008-

53015 20080321. Priority: US 2007-909315 20070330; US 2007-982309 20071024. CAN

152:144974 AN 2010:85102 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Disclosed herein are nucleoside phosphoramidates prodrugs I, wherein R and R3 are independently H,

alkyl, cycloalkyl, alkylamine, hydroxyalkyl, CH2SH, alkyl-sulfonyl, (CH2)3NHC(=NH)NH2, (1H-

indol-3-yl)methyl, (1H-imidazol-4-yl)methyl, acyl, aryl, aryl-alkyl; R3 and R both are alkyl; R3 and R

together are alkylidene so as to form a spiro ring; R3 is H and R and R2 together are (CH2)n so as to

form a cyclic ring that includes the adjoining N and C atoms; R is H and R3 and R2 together are

(CH2)n. so as to form a cyclic ring; that includes the adjoining N and C atoms, n is 2 to 4; one of R

and R3 is H and the other is R3 CH3, Et, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2Ph,

CH2-indol-3-yl, -CH2CH2SCH3, CH2CO2H, CH2C(O)NH2, CH2CH2COOH, CH2CH2C(O)NH2,

CH2CH2CH2CH2NH2, -CH2CH2CH2NHC(NH)NH2, CH2-imidazol-4-yl, CH2OH, CH(OH)CH3,

53

Page 54: dengue

CH2((4'-OH)-Ph), CH2SH, cycloalkyl; R1 is H, alkyl, cycloalkyl, aryl; R2 is H, alkyl; R, R2 and R3

together are (CH2)n; R4 is H, alkyl, alkoxy, alkylamino, halo, halo-alkyl, cycloalkyl, aminoacyl, aryl,

heterocycle; R5 is H, alkyl, CN, vinyl, hydroxy-alkyl, CH2OH, CH2F, CH2CN, CH2NH2,

CH2NHMe, CH2NMe2, alkyne; R6 is H, Me, CH2F, CHF2, CF3, F, CN; X is H, OH, F, OMe,

halogen, NH2, N3; Y is OH, H, alkyl, alkenyl, alkynyl, vinyl, N3, CN, halo, oxycarbonyl sulfonyl,

were prepd. and tested as antiviral agents. Thus, nucleoside II was prepd. and tested as antiviral agent

for the treatment of any condition the result of an infection by hepatitis C virus, West Nile virus,

yellow fever virus, dengue virus, rhinovirus, polio virus, hepatitis A virus, bovine viral diarrhea virus

or Japanese encephalitis virus.

18- Method to control dengue viruses in humans by picolinic acid and derivatives thereof.

Fernandez-Pol, Jose Alberto; Fernandez-Pol, Sebastian. (USA). U.S. Pat. Appl. Publ. (2010),

21pp. CODEN: USXXCO US 2010015174 A1 20100121 Patent written in English. Application:

US 2008-175277 20080717. Priority: . CAN 152:160987 AN 2010:84916 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20100015174 A1 20100121 US 2008-175277 20080717

AR 72542 A1 20100908 AR 2009-102484 20090702

Priority Application

US 2008-175277 A 20080717

Abstract

A method treats and then prevents a virus for afflicting an animal or a human as a metalloprotein

mediates the virus. The method administers systemically a therapeutic pharmacol. agent of picolinic

acid either singly or with interferons, chemokines or cytokines to fight dengue fever virus. The

picolinic acid inactivates the metalloprotein that allows replication of the virus. The invention also

includes the use of picolinic acid derivs. The viral proteins disintegrate by macrophage proteolytic

enzymes stimulated by the picolinic acid.

54

Page 55: dengue

2009 – 22 patents

1- Compositions and methods for Dengue virus (DV) treatment and vaccination. Shresta,

Sujan; Yauch, Lauren E.; Sette, Alessandro. (La Jolla Institute for Allergy and Immunology, USA).

PCT Int. Appl. (2009), 77 pp. CODEN: PIXXD2 WO 2009152147 A2 20091217 Designated

States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CL, CN,

CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR,

HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD,

ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL, PT, RO, RS,

RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM. Designated States RW: AT, BE, CH, CY, DE,

DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM,

GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2009-US46740

20090609. Priority: US 2008-60088 20080609. CAN 152:72884 AN 2009:1570230 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention relates to Dengue virus peptides and compns. thereof, and methods that employ Dengue

virus peptides and compns. thereof. The peptides include a portion of Dengue virus structural (core,

membrane or envelope) or non-structural (NS1, NS2A, NS2B, NS3, NS4A, NS4B or NS5)

polypeptide sequence eliciting a CD8+ T cell response against a DENV1, DENV2, DENV3 or

DENV4 serotype. The invention includes among other things, methods of treating Dengue virus

infection or pathol., which include, for example, administering Dengue virus peptide T cell epitope, to

treat a Dengue virus infection or pathol. The invention includes among other things Dengue virus

vaccination and immunization methods.

2- Preparation of nucleoside cyclic phosphates as antiviral agents. Du, Jinfa; Nagarathnam,

Dhanapalan; Pamulapati, Ganapati Reddy; Ross, Bruce S.; Sofia, Michael Joseph. (Pharmasset, Inc.,

USA). PCT Int. Appl. (2009), 145pp.; Chemical Indexing Equivalent to 152:381685 (US).

CODEN: PIXXD2 WO 2009152095 A2 20091217 Designated States W: AE, AG, AL, AM, AO,

AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM,

DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG,

KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX,

MY, MZ, NA, NG, NI, NO, NZ, OM, PE, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL,

SM, ST, SV, SY, TJ, TM. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE,

IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG.

Patent written in English. Application: WO 2009-US46619 20090608. Priority: US 2008-60683

55

Page 56: dengue

20080611; US 2008-140369 20081223; US 2009-479075 20090605. CAN 152:57501 AN

2009:1565702 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Cyclic phosphate of nucleoside derivs. I, wherein R1 is H, alkyl, cycloalkyl, alkaryl, aryl, halo-alkyl,

halo-alkenyl, halo-alkynyl, alkoxy, halo-alkoxy, CO2H, carboxylate, CONH2, substituted amide,

CH=CHCO2H, CH=CH-carboxylate; R2 is H, alkyl, CN, Me, vinyl, O-alkyl, OMe, OEt, hydroxy-

alkyl, CHwF, N3, CH2CN, CH2N3, CH2NH2, CH2NHMe, CH2NMe2, ethynyl-alkyne, halogen; R3

is H, Me, CH2F, CHF2, CF3, F, CN; X is H, OH, F, OMe, halogen, NH2, N3; B is nucleobase; were

prepd. and used for the treatment of viral infections in mammals, which is a compd., its stereoisomers,

salts (acid or basic addn. salts), hydrates, solvates. Thus, nucleotide II was prepd. and used as antiviral

agent.

3- Application of palmatine in preparing medicine for treating dengue virus infection. Yuan,

Zhiming; Jia, Fan; Zou, Gang; Li, Jing; Shi, Peiyong; Zheng, Dasheng; Cai, Quanxin; Yan, Jianping.

(Wuhan Institute of Virology, Chinese Academy of Sciences, Peop. Rep. China). Faming Zhuanli

Shenqing Gongkai Shuomingshu (2009), 10pp. CODEN: CNXXEV CN 101596192 A

20091209 Patent written in Chinese. Application: CN 2009-10063164 20090714. Priority: . CAN

152:67596 AN 2009:1551932 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

CN 101596192 A 20091209 CN 2009-10063164 20090714

Priority Application

CN 2009-10063164 20090714

Abstract

The invention relates to application of palmatine in prepg. medicine for treating or preventing dengue

virus infection. Palmatine can effectively inhibit dengue virus and has little cytotoxicity.

4- PIP-2 inhibition-based antiviral and anti-hyperlipidemic therapies. Glenn, Jeffrey S.; Cho,

Nam-Joon; Pang, Phillip S.; Lee, Choongho. (Leland Stanford Junior University, USA). PCT Int.

Appl. (2009), 58pp. CODEN: PIXXD2 WO 2009148541 A1 20091210 Designated States W:

AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU,

CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL,

56

Page 57: dengue

IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG,

MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE,

SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW: AT, BE, CH, CY, DE, DK, ES,

FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML,

MR, NE, SN, TD, TG. Patent written in English. Application: WO 2009-US3271 20090529.

Priority: US 2008-57188 20080529. CAN 152:1807 AN 2009:1536387 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Abstract

Interaction of a specific viral domain with phosphatidylinositol 4,5-bisphosphate (PIP 2) is shown to

mediate viral replication. Basic Amino Acid PIP-2 Pincer (BAAPP) domains are described herein,

including, without limitation, NS5A protein of HCV, NS4B protein of HCV, poliovirus, and

rhinovirus.

5- Enhanced antiviral therapy methods and devices comprising lectin affinity hemodialysis.

Tullis, Richard H.; Handley, Harold H., Jr.; Duffin, R. Paul; Joyce, James A. (Aethlon Medical Inc.,

USA). PCT Int. Appl. (2009), 60pp. CODEN: PIXXD2 WO 2009149179 A2 20091210

Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA,

CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM,

GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU,

LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT,

RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN. Designated States RW: AT, BE,

CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF,

CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2009-

US46123 20090603. Priority: US 2008-58536 20080603. CAN 152:27282 AN 2009:1536373

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Embodiments of the present invention relate to enhanced antiviral therapy methods, devices, and kits

for treating viral infections. The disclosed enhanced antiviral therapy methods, devices, and kits

enhance the efficacy of an antiviral therapy by administering a lectin affinity hemodialysis treatment

to an individual suffering from viral infection in combination with the antiviral therapy. A patient

suffering from HCV infection was administered a lectin affinity hemodialysis treatment for 8 h a day,

3 times a week for 12 wk. The viral load of the patient reduced significantly compared to the

administration of the interferon/ribavirin therapy alone, and the patient achieved a sustained viral

response after 12 wk of lectin affinity hemodialysis treatment.

57

Page 58: dengue

6- Small molecule inhibitors for the treatment or prevention of dengue virus and other viral

infection. Byrd, Chelsea M.; Jordan, Robert; Dai, Dongcheng; Hruby, Dennis E. (Siga

Technologies, Inc., USA). PCT Int. Appl. (2009), 88pp. CODEN: PIXXD2 WO 2009149054

A1 20091210 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW,

BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD,

GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR,

LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG,

PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN. Designated States

RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR,

BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application:

WO 2009-US45921 20090602. Priority: US 2008-58263 20080603. CAN 152:27275 AN

2009:1532703 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Methods and pharmaceutical compns. for treating viral infections, by administering certain compds. in

therapeutically effective amts. are disclosed. Methods of using the compds. and pharmaceutical

compns. thereof are also disclosed. In particular, the treatment and prophylaxis of viral infections

such as caused by flavivirus is disclosed, i.e., including but not limited to, Dengue virus, West Nile

virus, yellow fever virus, Japanese encephalitis virus, and tick-borne encephalitis virus. A compd.

library was screened for inhibitory activity against dengue virus-induced cytopathic effect on Vero

cells. Active compds. were tested for activity in viral yield assays carried out at several drug concns.

I was one of the most potent and selective compds. from the pool of initial 22 quality hits, with

activity against all four serotypes of dengue virus. Hard gelatin capsules contain active ingredient

30.0, starch 305.0, and magnesium stearate 5.0 mg/capsule.

7- Preparation of 1'-substituted carba-nucleoside analogs as antiviral agents. Butler, Thomas;

Cho, Aesop; Kim, Choung U.; Saunders, Oliver L.; Zhang, Lijun. (Gilead Sciences, Inc., USA).

PCT Int. Appl. (2009), 173pp. CODEN: PIXXD2 WO 2009132135 A1 20091029 Designated

States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO,

CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU,

ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME,

MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC,

SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW: AT, BE, CH, CY, DE,

DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM,

GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2009-US41447

58

Page 59: dengue

20090422. Priority: US 2008-47263 20080423; US 2008-139449 20081219. CAN 151:470463

AN 2009:1325388 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

1'-Substituted carba-nucleoside analogs I, wherein R1-R5 are independently H, OR, NR2, N3, CN,

NO2, S(O)nR, halogen, alkyl, carbocyclyl-alkyl, alkenyl, alkynyl, arylalkyl; any two of R1-R5 on

adjacent carbo when taken together are O(CO)O or when taken together with the ring carbon to which

they are attached form a double bond; R6 is id OR, NR2, N3, CN, NO2, S(O)nR, acyl, carboxylate,

amide, thio ester, sulfonyl, sulfonamide, halogen, alkyl, carbocyclyl-alkyl, alkylalkenyl, alkenyl,

alkynyl, arylalkyl; R1R6 or R2R6 together form O(CO)O; n is 0-2; R is H, alkyl, alkenyl, alkynyl,

arylalkyl, carbocyclyl-alkyl, acyl, carboxylate ester, amide, thio ester, sulfonyl, sulfonamide; R7 is H,

acyl, carboxylate ester, amide, sulfonyl, sulfonamide, substituted phosphorus; R8 is halogen,

substituted N, N3, NO, NO2, CHO, CN, imine, oxime, ketal, amide, sulfonamide, carboxylate ester,

alkyl, alkenyl, alkynyl, carbocycloalkyl; R9 and R10 are independently H, halogen, substituted N, N3,

NO, substituted hydrazine, NO2, CHO, CN, imine, oxime, ketal, amide, thioamide, carboxylate ester,,

were prepd. and used as antiviral agents. The compds., compns., and methods provided are useful for

the treatment of Flaviviridae virus infections, particularly hepatitis C infections. Thus, nucleoside II

was prepd. and tested as antiviral agent (EC50 10-100 μM), wherein the viral infection is caused by a

virus selected from the group consisting of dengue virus, yellow fever virus, West Nile virus, Japanese

encephalitis virus, tick-borne encephalitis virus, Kunjin virus, Murray Valley encephalitis virus, St.

Louis encephalitis virus, Omsk hemorrhagic fever virus, bovine viral diarrhea virus, Zika virus and

Hepatitis C virus.

8- Preparation of 1'-substituted carba-nucleoside analogs as antiviral agents carba-nucleoside

analogs for antiviral treatment. Cho, Aesop; Kim, Choung U.; Parrish, Jay; Xu, Jie. (Gilead

Sciences, Inc., USA). PCT Int. Appl. (2009), 154pp. CODEN: PIXXD2 WO 2009132123 A1

20091029 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH,

GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT,

LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2009-US41432 20090422. Priority: US 2008-47263 20080423; US 2008-139449 20081219. CAN

151:491358 AN 2009:1325387 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

59

Page 60: dengue

Abstract

1'-Substituted carba-nucleoside analogs I, wherein R1-R5 are independently H, OR, NR2, N3, CN,

NO2, S(O)nR, halogen, alkyl, carbocyclyl-alkyl, alkenyl, alkynyl, arylalkyl; any two of R1-R5 on

adjacent carbo when taken together are O(CO)O or when taken together with the ring carbon to which

they are attached form a double bond; R6 is id OR, NR2, N3, CN, NO2, S(O)nR, acyl, carboxylate,

amide, thio ester, sulfonyl, sulfonamide, halogen, alkyl, carbocyclyl-alkyl, alkylalkenyl, alkenyl,

alkynyl, arylalkyl; R1R6 or R2R6 together form O(CO)O; n is 0-2; R is H, alkyl, alkenyl, alkynyl,

arylalkyl, carbocyclyl-alkyl, acyl, carboxylate ester, amide, thio ester, sulfonyl, sulfonamide; R7 is H,

acyl, carboxylate ester, amide, sulfonyl, sulfonamide, substituted phosphorus; R8 is halogen,

substituted N, N3, NO, NO2, CHO, CN, imine, oxime, ketal, amide, sulfonamide, carboxylate ester,

alkyl, alkenyl, alkynyl, carbocycloalkyl; R9 and R10 are independently H, halogen, substituted N, N3,

NO, substituted hydrazine, NO2, CHO, CN, imine, oxime, ketal, amide, thioamide, carboxylate ester,,

were prepd. and used as antiviral agents. The compds., compns., and methods provided are useful for

the treatment of Flaviviridae virus infections, particularly hepatitis C infections. Thus, nucleoside II

was prepd. and tested as antiviral agent (EC50 10-100 μM), wherein the viral infection is caused by a

virus selected from the group consisting of dengue virus, yellow fever virus, West Nile virus, Japanese

encephalitis virus, tick-borne encephalitis virus, Kunjin virus, Murray Valley encephalitis virus, St.

Louis encephalitis virus, Omsk hemorrhagic fever virus, bovine viral diarrhea virus, Zika virus and

Hepatitis C virus.

9- Chemical compounds having antiviral activity against dengue virus and other flaviviruses.

Mazola Reyes, Yuliet; Chinea Santiago, Glay; Guirola Cruz, Osmany; Vera Alvarez, Roberto; Huerta

Galindo, Vivian; Fleitas Salazar, Noralvis; Musacchio Lasa, Alexis. (Centro de Ingenieria Genetica y

Biotecnologia, Cuba). PCT Int. Appl. (2009), 81pp. CODEN: PIXXD2 WO 2009106019 A2

20090903 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH,

GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT,

LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in Spanish. Application: WO

2009-CU2 20090227. Priority: CU 2008-28 20080229. CAN 151:280183 AN 2009:1082068

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

60

Page 61: dengue

Abstract

The invention relates to the use of chem. compds. obtained in silico for the prepn. of pharmaceutical

compns. for attenuating or inhibiting dengue virus infection, in particular by interference with or

modulation of the multiple steps of the viral replication cycle assocd. with the arrival of the virus in

the target cells and the assembly of the progeny virions. The invention also relates to the use of said

pharmaceutical compns. for the prophylactic or therapeutic treatment of infection caused by the four

dengue virus serotypes and by other flaviviruses. The compds. are composed of two functional

subunits denoted, [C]-[A]. Where [A] is an anchor capable of latching on to the lj loop of protein E.

The anchor may be a hydrogen donor, a hydrogen acceptor, a hydrophobic element, a hydrogen donor

and acceptor simultaneously. The "head" subunit, or [C], contains the antiviral activity and it is

covalently attached to subunit [A].

10- Development of a broad-spectrum antiviral with activity against Ebola virus. Aman, M.

Javad; Kinch, Michael S.; Warfield, Kelly; Warren, Travis; Yunus, Abdul; Enterlein, Sven; Stavale,

Eric; Wang, Peifang; Chang, Shaojing; Tang, Qingsong; Porter, Kevin; Goldblatt, Michael; Bavari,

Sina. United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD,

USA. Antiviral Research (2009), 83(3), 245-251. Publisher: Elsevier B.V., CODEN: ARSRDR

ISSN: 0166-3542. Journal written in English. CAN 151:417551 AN 2009:1001305 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

We report herein the identification of a small mol. therapeutic, FGI-106, which displays potent and

broad-spectrum inhibition of lethal viral hemorrhagic fevers pathogens, including Ebola, Rift Valley

and Dengue Fever viruses, in cell-based assays. Using mouse models of Ebola virus, we further

demonstrate that FGI-106 can protect animals from an otherwise lethal infection when used either in a

prophylactic or therapeutic setting. A single treatment, administered 1 day after infection, is sufficient

to protect animals from lethal Ebola virus challenge. Cell-based assays also identified inhibitory

activity against divergent virus families, which supports a hypothesis that FGI-106 interferes with a

common pathway utilized by different viruses. These findings suggest FGI-106 may provide an

opportunity for targeting viral diseases.

11- Methods of inhibiting viral infection. Kinch, Michael; Goldblatt, Michael. (Functional

Genetics, Inc., USA). PCT Int. Appl. (2009), 65pp. CODEN: PIXXD2 WO 2009091435 A2

20090723 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH,

GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT,

61

Page 62: dengue

LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2008-US81904 20081031. Priority: US 2007-983966 20071031. CAN 151:189938 AN

2009:885111 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Compds., pharmaceutical compns. and methods of inhibiting viral infection in a mammal in need of

same, are provided, which employ quinoline compds. This family of compds., designated FGI-104

herein, inhibits viral infection therapeutically and prophylactically.

12- An astragalus extract as an antiviral for several genera of the flaviviridae family. Zhong,

Shouming; Yu, Hongwen; Blair, Edward. (Phynova Limited, UK). PCT Int. Appl. (2009), 89pp.;

Chemical Indexing Equivalent to 151:16632 (GB). CODEN: PIXXD2 WO 2009068872 A1

20090604 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH,

GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT,

LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2008-GB3947 20081126. Priority: GB 2007-23609 20071127. CAN 151:16633 AN 2009:669997

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention relates to a novel antiviral product and its use in the treatment of the flaviviridae

family of viruses including the genus flavivirus, particularly Dengue. An antiviral product and its use

in the treatment of the Flaviviridae family of viruses including the genera Flavivirus, particularly

Dengue, Pestivirus, Unassigned Flaviviradae, and tentative Species of the Genus HCV. The antiviral

product is an ext. of Astragalus and preferably comprises at least one marker selected from

astragaloside I, astragaloside IV, formononetin-7-O-β-D-glucoside and 3'-hydroxyl-formononetin-7-O-

β-D-glucoside.

62

Page 63: dengue

13- Dengue virus 2 non-structural protein 2b for preparing DNA vaccines, antibodies, anti-

dengue virus agents and NS2B research. Liu, Limei; An, Jing; Chen, Zongtao. (Third Military

Medical University, PLA, Peop. Rep. China). Faming Zhuanli Shenqing (2009), 24pp. CODEN:

CNXXEV CN 101429236 A 20090513 Patent written in Chinese. Application: CN 2008-

10233216 20081203. Priority: . CAN 151:31759 AN 2009:596452 CAPLUS (Copyright (C)

2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

CN 101429236 A 20090513 CN 2008-10233216 20081203

Priority Application

CN 2008-10233216 20081203

Abstract

The dengue virus type 2 non-structural protein 2B (NS2B) has an amino acid sequence of SEQ ID

No.2. The invention also provides a recombination expression vector and a transformant contg.

NS2B. The recombination expression vector successfully exhibits NS2B in eukaryotic cell, induces

specific anti-NS2B polyclonal antibody in mouse, and can be used for prepg. nucleic acid vaccine

against dengue virus 2 type and anti-NS2B antibody.

14- Antiviral agents containing silver-chloro complex. Yokosawa, Hirotsugu. (Japan). Jpn.

Kokai Tokkyo Koho (2009), 15pp. CODEN: JKXXAF JP 2009096745 A 20090507 Patent

written in Japanese. Application: JP 2007-268414 20071015. Priority: . CAN 150:488982 AN

2009:549478 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

JP 2009096745 A 20090507 JP 2007-268414 20071015

Priority Application

JP 2007-268414 20071015

63

Page 64: dengue

Abstract

The antiviral agents and antiviral method whose effect is maintained, are developed. The antiviral

agents of this invention contains the silver chloro complex so that it may be supplied by the silver

concn. of 2.4-40 mg/L. Surprisingly, it was discovered that when the silver concn. of the silver-chloro

complex soln. is much lower than the concn. range usually used as antibacterial agents (100 - 200

mg/L), it can actualize virus control.

15 - Optimized dengue virus entry inhibitory peptide (DN81). Michael, Scott F.; Isern, Sharon;

Garry, Robert; Samudrala, Ram; Costin, Joshua; Jenwitheesuk, Ekachai. (Florida Gulf Coast

University, USA). PCT Int. Appl. (2009), 19pp. CODEN: PIXXD2 WO 2009048658 A2

20090416 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH,

GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT,

LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2008-US69716 20080711. Priority: US 2007-949710 20070713. CAN 150:414217 AN

2009:456348 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention provides peptide entry inhibitors bindable to regions in viral class II E proteins. The

interaction of an inhibitor with such regions, or the modulation of the activity of such regions with an

inhibitor, could inhibit viral fusion and hence viral infectivity. In one aspect, the invention provides

compds. and methods of screening the compds. against these bindable regions in order to discover

therapeutic candidates for a disease caused by a virus having a class II protein. Furthermore, the

invention provides for methods of inhibiting viral infection by dengue virus and/or binding between

the virion envelope of dengue viruses and membranes of the target cell (the process that delivers the

viral genome into the cell cytoplasm). The invention provides for methods that employ peptides or

peptide derivs. to inhibit dengue virus:cell binding. The invention provides for methods of treatment

of diseases induced by the dengue virus. The DN81 peptide showed an increase in inhibitory activity

as a function of concn.

15- Optimized dengue virus entry inhibitory peptide (10AN). Michael, Scott F.; Isern, Sharon;

Costin, Joshua; Samudrala, Ram; Jenwitheesuk, Ekachai. (Florida Gulf Coast University, USA).

PCT Int. Appl. (2009), 19pp. CODEN: PIXXD2 WO 2009045596 A2 20090409 Designated

64

Page 65: dengue

States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO,

CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU,

ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME,

MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC,

SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW: AT, BE, CH, CY, DE,

DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM,

GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2008-US69725

20080711. Priority: US 2007-949733 20070713. CAN 150:431673 AN 2009:424610 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention relates to peptide entry inhibitors and methods of detg. such inhibitors that are bindable

to regions of viruses having class II E proteins, such as the dengue virus E protein, as candidates for in

vivo anti-viral compds. Thus, the 10AN peptide having a SEQ ID NO 1:

FWFTLIKTQAKQPARYRRFC showed an increase in inhibitory activity against dengue virus 2

(DENV-2) in vitro as a function of concn., as demonstrated using focus-forming assays.

16- microRNA profiles in the diagnosis of viral infection and the identification of microRNAs as

targets for antiviral therapy. Kowalik, Timothy F.; Stadler, Bradford M. (University of

Massachusetts, USA). PCT Int. Appl. (2009), 194pp. CODEN: PIXXD2 WO 2009033185 A1

20090312 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY,

BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH,

GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT,

LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ,

CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO

2008-US75646 20080908. Priority: US 2007-967780 20070906. CAN 150:304161 AN

2009:292432 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention related to miRNA signatures and diagnostic and therapeutic applications of

miRNA signatures. The miRNA signatures are defined by a test sample miRNA profile relative to an

appropriate control miRNA profile. In some embodiments, the test sample is a sample isolated,

obtained or derived from a virus- infected cell or organism. The present invention further relates to

the use of miRNA signatures in the identification of draggable targets and antiviral agents. Kits and

65

Page 66: dengue

compns. are also provided. The identification of specific miRNA signatures for infection of human

cell lines by adenovirus 5 and human cytomegalovirus.

17- Preparation of acridinone derivatives as antiparasitic, antifungal and antiviral agents.

Pellon Condom, Rolando F.; Docampo Palacios, Maite L.; Pardo Andreu, Gilberto L.; Fernandez

-Calienes Valdes, Ayme; Mendiola Martinez, Barbara Judith; Rojas Rivero, Lazara; D'Accorso Haicck,

Norma Beatriz; Fascio Silva, Mirta Liliana; Damonte Lotito, Elsa Beatriz; Garcia Cattebeke, Cybele

Carina; Sepulveda Suchecki, Claudia Soledad; Mazzucco Gavieiro, Maria Belen; Talarico Salinas,

Laura Beatriz; Maes, Louis. (Centro de Quimica Farmaceutica, Cuba). PCT Int. Appl. (2009),

40pp. CODEN: PIXXD2 WO 2009026858 A1 20090305 Designated States W: AE, AG, AL, AM,

AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM,

DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG,

KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX,

MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM,

ST, SV, SY, TJ, TM, TN, TR. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR,

IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD,

TG. Patent written in Spanish. Application: WO 2008-CU7 20080828. Priority: CU 2007-202

20070828. CAN 150:283334 AN 2009:259420 CAPLUS (Copyright (C) 2010 ACS on SciFinder

(R))

Abstract

The invention relates to the synthesis and evaluation of novel acridinone derivs. as antiparasitic,

antifungal and antiviral agents. More specifically, the invention relates to 10-allyl-, 10-(3-methyl-2-

butenyl)-, 10-(1,2-propanodienyl)-9(10H)-acridinone, and 10-[[3-[(4R,5R,6S,7R)-6-hydroxy-2,2-

dimethyltetrahydrofuro[2,3-d][1,3]dioxol-5-yl]-4,5-dihydro-5-isoxazolyl]methyl]-9(10H)acridinone

(I) or their derivs. in which positions 1-8 of the acridinone ring may be substituted by halogen,

hydroxy, amino, nitro, alkyl, alkoxy, and other groups. The acridinones have an activity against

Plasmodium falciparum, Trypanosoma brucei and Trypanosoma cruzi protozoans, as well as

Microsporum canis and strains of the Junin virus and Dengue. Thus, acridinone deriv. I was prepd.

from 9-acridinone by allylation followed by cycloaddn. reaction with 1,2-O-isopropylidene-α-D-

xylopentadialdo-1,4-furanose oxime. The possible biol. mode of action of the acridinone derivs. was

studied using 10-allyl-6-chloro-2-fluoro-9(10H)-acridinone as model compd.

66

Page 67: dengue

18- TRAIL antigens in dengue fever and their use as targets in treating dengue virus infection.

Bosch, Irene; Warke, Rajas V.; Martin, Katherine J. (University of Massachusetts Medical School,

USA). PCT Int. Appl. (2009), 145pp. CODEN: PIXXD2 WO 2009025743 A2 20090226

Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA,

CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT,

HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY,

MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO,

RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM, TN, TR. Designated States RW: AT, BE,

CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF,

CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2008-

US9666 20080813. Priority: US 2007-965173 20070817. CAN 150:275521 AN 2009:238890

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

TRAIL antigens are shown to play a role in the development of dengue fever, an important tropical

illness for which there is currently no virus-specific treatment, and may therefore be useful in the

treatment of the disease. Gene expression profiling in cultured primary human cells, including human

umbilical vein endothelial cells (HUVECs), dendritic cells (DCs), monocytes and B cells, using the

com. Affymetrix GeneChip (HG-U1 33A), identified changes in response to dengue virus infection.

Tissue necrosis factor-related apoptosis inducing ligand (TRAIL) gene expression was a common

response in all cells and may play a role as an immunomodulator in infected cells. Interactions

between type-I and II interferon response genes and TRAIL are expected to increase the innate

immunity to the virus or even to other pathogens like bacteria. Dengue virus induces TRAIL

expression in immune cells and HUVECs at the mRNA and protein level and was found to be

dependent on an intact interferon type I signaling pathway. Anti-TRAIL antibody incubation with

primary cells showed an increase in dengue virus accumulation and conversely, a decrease in viral

RNA when TRAIL was added to the culture. These data suggest that TRAIL may play a role in the

anti-viral response to dengue fever and that it is a candidate for anti-viral interventions to the disease.

Further, TRAIL antiviral function does not promote apoptosis. The role of exogenous TRAIL in

dendritic cells confirmed a strong anti-inflammatory response due to the lowering of prodn. of

mediators of inflammation present in dengue infection. TRAIL may also be useful in the treatment of

other flavivirus infections.

19- Iminosugar compounds with antiflavivirus activity. Gu, Baohua; Block, Timothy M.;

Moriarty, Robert M.; Deshpande, Mahendra N.; Shah, Rajendra C. (The Board of Trustees of the

University of Illinois, USA). U.S. Pat. Appl. Publ. (2009), 27 pp. CODEN: USXXCO US

67

Page 68: dengue

2009042268 A1 20090212 Patent written in English. Application: US 2008-112694 20080430.

Priority: US 2007-914889 20070430. CAN 150:229640 AN 2009:172500 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20090042268 A1 20090212 US 2008-112694 20080430

Priority Application

US 2007-914889P P 20070430

Abstract

An anti-viral compds. effective against viruses belonging to the Flaviviridae family, wherein the anti-

viral compds. are 1,5-dideoxy-1,5-imino-D-glucitol deriv. compds.

20- Dengue virus nonstructural protein NS1-based methods for identifying agents for treating

dengue hemorrhagic fever. Ikuta, Kazuyoshi; Kurosu, Takeshi; Anantapreecha, Surapee;

Sawanpanyalert, Pathom. (Osaka University, Japan; National Institute of Health (NIH), Department of

Medical Sciences (DMSC); Ministry of Public Health, Nonthaburi). PCT Int. Appl. (2009), 45

pp. CODEN: PIXXD2 WO 2009016831 A1 20090205 Designated States W: AE, AG, AL, AM,

AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM,

DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG,

KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX,

MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM,

ST, SV, SY, TJ, TM, TN, TR. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR,

IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD,

TG. Patent written in English. Application: WO 2008-JP2035 20080730. Priority: US 2007-

953583 20070802. CAN 150:183353 AN 2009:140200 CAPLUS (Copyright (C) 2010 ACS on

SciFinder (R))

Abstract

Compds. that inhibit the binding of dengue virus nonstructural protein NS1 to complement regulatory

protein clusterin, or that inhibit complement activation by NS1, can be used for treating dengue virus

infection, dengue hemorrhagic fever, and dengue shock syndrome. Such compds. can be identified

using methods that measure inhibition of NS1/clusterin binding, and/or inhibition of complement

activation.

68

Page 69: dengue

21- Fc variants of monoclonal antibodies against dengue and other viruses. Goncalvez, Ana P.;

Purcell, Robert H.; Lai, Ching-Juh. (United States Dept. of Health & Human Services, USA). PCT

Int. Appl. (2009), 95 pp. CODEN: PIXXD2 WO 2009011941 A2 20090122 Designated States

W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR,

CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID,

IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG,

MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE,

SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT. Designated States RW: AT, BE, CH, CY, DE, DK, ES,

FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML,

MR, NE, SN, TD, TG. Patent written in English. Application: WO 2008-US59313 20080403.

Priority: US 2007-922282 20070404; US 2007-927755 20070504; US 2007-928405 20070508.

CAN 150:166232 AN 2009:93361 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The authors disclose Fc region variants of antibodies the bind an Fcγ receptor (FcγR) with lower

affinity than the parent antibodies. The variants, targeting the envelope glycoprotein of dengue virus,

are shown to exhibit reduced or absent antibody-dependent enhancement of infection.

22- Antiviral properties of zosteric acid and related molecules. Michael, Scott F.; Isern, Sharon;

Costin, Joshua. (Florida Gulf Coast University, USA). PCT Int. Appl. (2009), 36pp. CODEN:

PIXXD2 WO 2009012157 A2 20090122 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ,

BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE,

EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR,

KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG,

NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TJ, TM,

TN, TR. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC,

MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written

in English. Application: WO 2008-US69808 20080711. Priority: US 2007-949694 20070713; US

2008-58026 20080602. CAN 150:136611 AN 2009:79032 CAPLUS (Copyright (C) 2010 ACS

on SciFinder (R))

Abstract

The invention relates chem. compd. entry inhibitors and methods of detg. such inhibitors that interact

with regions of viruses, such as the dengue virus, as candidates for in vivo antiviral compds. Compds.

of the invention include zosteric acid and related mols.

69

Page 70: dengue

2008 – 8 patents

1- Use of sequence covariance networks for identification of targets for designing antiviral

agents and for diagnostic applications. Aurora, Rajeev; Donlin, Maureen J.; Tavis, John E. (Saint

Louis University, USA). U.S. Pat. Appl. Publ. (2008), 23pp. CODEN: USXXCO US

2008318207 A1 20081225 Patent written in English. Application: US 2008-144030 20080623.

Priority: US 2007-945543 20070621; US 2007-987696 20071113. CAN 150:71091 AN

2008:1532900 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20080318207 A1 20081225 US 2008-144030 20080623

Priority Application

US 2007-945543P P 20070621

US 2007-987696P P 20071113

Abstract

Methods of identifying targets for designing a therapeutic agent are disclosed. These methods

comprise: detg. an amino acid sequence of one or more polypeptides of each isolate of a plurality of

isolates of a biol. system; identifying covariance pairs of amino acid residues; establishing a network

comprising the covariance pairs; and identifying one or more hub residue positions, wherein a hub

residue position comprises a target for designing a therapeutic agent if the hub residue position has a

rank order in the 40th percentile or greater. Methods are disclosed for selecting a therapy for an

infectious disorder, in particular methods for selecting an antiviral therapy. In various configurations,

these methods comprise: detg. amino acids occupying a plurality of diagnostic amino acid residue

positions comprised by one or more polypeptides encoded by an infectious agent infecting a subject;

and assigning the infectious agent infecting the subject to one covariance network selected from a

plurality of covariance networks, wherein each network comprises a unique rank order of hubs with

respect to the other networks, and whereby the therapy is selected on the basis of the covariance

network assignment. The invention also provides methods for designing antimicrobial and antiviral

therapeutic agents, and methods for establishing a differential diagnosis.

70

Page 71: dengue

2- Antiviral drugs for treatment or prevention of dengue infection. Byrd, Chelsea M.; Jordan,

Robert; Dai, Dongcheng; Bolken, Tove; Hruby, Dennis E. (Siga Technologies, Inc., USA). PCT Int.

Appl. (2008), 67pp. CODEN: PIXXD2 WO 2008147962 A1 20081204 Designated States W:

AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU,

CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL,

IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG,

MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE,

SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT. Designated States RW: AT, BE, CH, CY, DE, DK, ES,

FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML,

MR, NE, SN, TD, TG. Patent written in English. Application: WO 2008-US64662 20080523.

Priority: US 2007-924628 20070523. CAN 150:16036 AN 2008:1454478 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Abstract

Compds., methods and pharmaceutical compns. for treating viral infections, by administering certain

compds. in therapeutically effective amts. are disclosed. Methods for prepg. the compds. and methods

of using the compds. and pharmaceutical compns. thereof are also disclosed. In particular, the

treatment and prophylaxis of viral infections such as caused by flavivirus is disclosed, i.e., including

but not limited to, Dengue virus, West Nile virus, yellow fever virus, Japanese encephalitis virus, and

tick-borne encephalitis virus. The present invention provides a pharmaceutical compn. comprising a

pharmaceutically acceptable carrier and a compd. having the following general formula Ar-SO2-

NR1R2 wherein R1 and R2 are independently hydrogen, alkyl, alkenyl, alkynyl, or unsubstituted or

substituted cycloalkyl, arylalkyl, aryl, or R1 and R2 together may form a substituted or unsubstituted

ring, which may include one or more heteroatoms in the ring; and Ar is substituted or unsubstituted

aryl or heteroaryl.

3- Antiviral amphipathic helical peptides derived from HCV NS5A membrane anchoring

peptide. Chisari, Francis V. (The Scripps Research Institute, USA). PCT Int. Appl. (2008),

242pp. CODEN: PIXXD2 WO 2008133759 A2 20081106 Designated States W: AE, AG, AL, AM,

AO, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM,

DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG,

KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX,

MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM,

SV, SY, TJ, TM, TN, TR, TT. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR,

IE, IS, IT, LU, MC, MT, NL, NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD,

TG. Patent written in English. Application: WO 2008-US452 20080110. Priority: US 2007-

71

Page 72: dengue

879727 20070110; US 2007-967783 20070907. CAN 149:525347 AN 2008:1338066 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present application is directed to antiviral peptides, methods of using these peptides to prevent or

inhibit infections by a human immunodeficiency virus or a virus from the Flaviviridae family, and

pharmaceutical compns. and combinations, as well as articles of manuf. comprising these peptides. In

particular, an amphipathic alpha-helical peptide derived from the membrane anchor domain of the

Hepatitis C virus (HCV) NS5A protein is identified to inhibit HCV infection by inactivating both

extracellular and intracellular infectious particles. By screening its derived peptide library, more

peptide derivs. or analogs sharing eight consensus amino acid sequences are also identified to be

virocidal in broad range, to flaviviruses, paramyxoviruses, and human immunodeficiency virus. For

HIV, these antiviral peptides prevent its infection by disrupting the integrity of the viral membrane and

capsid core while preserving the integrity of host membranes.

4- Preparation of nucleoside phosphoramidate prodrugs as antiviral agents. Sofia, Michael J.;

Du, Jinfa; Wang, Peiyuan; Nagarathnam, Dhanapalan. (Pharmasset, Inc., USA). PCT Int. Appl.

(2008), 751 pp., Chemical Indexing Equivalent to 152:144974 (US). CODEN: PIXXD2 WO

2008121634 A2 20081009 Designated States W: AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG,

BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI,

GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC,

LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ,

OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL,

NO, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

Application: WO 2008-US58183 20080326. Priority: US 2007-909315 20070330; US 2007-

982309 20071024; US 2008-53015 20080321. CAN 149:426212 AN 2008:1215396 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Disclosed herein are nucleoside phosphoramidates prodrugs I, wherein R and R3 are independently H,

alkyl, cycloalkyl, alkylamine, hydroxyalkyl, CH2SH, alkyl-sulfonyl, (CH2)3NHC(=NH)NH2, (1H-

indol-3-yl)methyl, (1H-imidazol-4-yl)methyl, acyl, aryl, aryl-alkyl; R3 and R both are alkyl; R3 and R

together are alkylidene so as to form a spiro ring; R3 is H and R and R2 together are (CH2)n so as to

form a cyclic ring that includes the adjoining N and C atoms; R is H and R3 and R2 together are

(CH2)n. so as to form a cyclic ring; that includes the adjoining N and C atoms, n is 2 to 4; one of R

and R3 is H and the other is R3 CH3, Et, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2Ph,

72

Page 73: dengue

CH2-indol-3-yl, -CH2CH2SCH3, CH2CO2H, CH2C(O)NH2, CH2CH2COOH, CH2CH2C(O)NH2,

CH2CH2CH2CH2NH2, -CH2CH2CH2NHC(NH)NH2, CH2-imidazol-4-yl, CH2OH, CH(OH)CH3,

CH2((4'-OH)-Ph), CH2SH, cycloalkyl; R1 is H, alkyl, cycloalkyl, aryl; R2 is H, alkyl; R, R2 and R3

together are (CH2)n; R4 is H, alkyl, alkoxy, alkylamino, halo, halo-alkyl, cycloalkyl, aminoacyl, aryl,

heterocycle; R5 is H, alkyl, CN, vinyl, hydroxy-alkyl, CH2OH, CH2F, CH2CN, CH2NH2,

CH2NHMe, CH2NMe2, alkyne; R6 is H, Me, CH2F, CHF2, CF3, F, CN; X is H, OH, F, OMe,

halogen, NH2, N3; Y is OH, H, alkyl, alkenyl, alkynyl, vinyl, N3, CN, halo, oxycarbonyl sulfonyl,

were prepd. and tested as antiviral agents. Thus, nucleoside II was prepd. and tested as antiviral agent

for the treatment of any condition the result of an infection by hepatitis C virus, West Nile virus,

yellow fever virus, dengue virus, rhinovirus, polio virus, hepatitis A virus, bovine viral diarrhea virus

or Japanese encephalitis virus.

5- Method using pigs for the evaluation of dengue virus antiviral agents and vaccines.

Burgess, Timothy H.; Porter, Kevin R.; Freilich, Daniel A.; Doolan, Denise L. (USA). U.S. Pat.

Appl. Publ. (2008), 10pp. CODEN: USXXCO US 2008219930 A1 20080911 Patent written in

English. Application: US 2006-507322 20060821. Priority: US 2005-709804 20050822. CAN

149:299778 AN 2008:1098279 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20080219930 A1 20080911 US 2006-507322 20060821

Priority Application

US 2005-709804P P 20050822

Abstract

The invention discloses a method for evaluating potential compds. and vaccines for the prevention or

treatment of dengue virus infection. The method utilizes pigs as an animal model for the evaluation of

test vaccine or drug compds. The breeds that can be used include Yorkshire or Lancashire, as well as

miniature pig breeds.

6- A dengue reporter virus and methods of producing and using it to identify dengue virus

inhibitors. Puffer, Bridget; Doranz, Benjamin J. (Integral Molecular, Inc., USA). PCT Int. Appl.

(2008), 57 pp. CODEN: PIXXD2 WO 2008051266 A2 20080502 Designated States W: AE,

AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK,

73

Page 74: dengue

DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG,

KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, ME, MG, MK, MN, MW,

MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL,

SM, SV, SY, TJ, TM, TN, TR, TT, TZ, UA. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI,

FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN,

TD, TG. Patent written in English. Application: WO 2007-US3660 20070212. Priority: US

2006-772916 20060213. CAN 148:487140 AN 2008:529145 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Abstract

The invention relates to the prodn. and uses of dengue virus replicons and dengue reporter virus

particles. The invention relates to methods for identifying inhibitors of dengue virus infection,

inhibitors of dengue virus replication, and inhibitors of dengue virus assembly.

7- Preparation of D-glucopyranose 1-[3,5-bis(1,1-dimethylethyl)-4-hydroxybenzoate] as antiviral

agent. Vachy, Robert. (RDW Pharma, Fr.). PCT Int. Appl. (2008), 18pp. CODEN: PIXXD2

WO 2008000920 A1 20080103 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG,

BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD,

GE, GH, GM, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS,

LT, LU, LV, LY, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL,

PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT,

SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in French.

Application: WO 2006-FR1486 20060623. Priority: . CAN 148:79261 AN 2008:12161

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

D-glucopyranose 1-[3,5-bis(1,1-dimethyl-ethyl)-4-hydroxybenzoate] was prepd. by glycosylation of

3,5-bis(t-Bu)-4-hydroxybenzoic acid with D-glucose. It applies in particular to the prepn. and the use

of this compd. and of its derivs. for the prepn. of medicaments for the treatment and/or prevention of

infections with enveloped viruses, and in particular, in humans, the Herpes virus, the AIDS virus, the

flu virus, the hepatitis B virus, the hepatitis C virus, the dengue virus and the Ebola virus.

8- Mitochondrial antiviral signaling protein in the prevention and treatment of viral diseases.

Chen, Zhijian; Bhargava, Rashu Seth; Sun, Lijun; Li, Xiao-Dong. (Board of Regents, The University

of Texas System, USA). U.S. Pat. Appl. Publ. (2008), 93pp. CODEN: USXXCO US

2008003614 A1 20080103 Patent written in English. Application: US 2006-509924 20060825.

74

Page 75: dengue

Priority: US 2005-711451 20050825. CAN 148:142884 AN 2008:9095 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20080003614 A1 20080103 US 2006-509924 20060825

US 7625724 B2 20091201

US 20100137562 A1 20100603 US 2009-628509 20091201

Priority Application

US 2005-711451P P 20050825

US 2006-509924 A3 20060825

Abstract

The present invention includes the identification and characterization of a novel protein designated

mitochondrial antiviral signaling (MAVS) that is essential for NF-κB and IRF3 activation by RNA

viruses. MAVS includes an N-terminal CARD-like domain and a C-terminal transmembrane domain

that targets the protein to the mitochondrial membrane. MAVS functions downstream of RIG-1 and

upstream of IκB and IRF3 phosphorylation. Suppression of MAVS expression blocks interferon

prodn. and exacerbates the viral replication and killing of the host cells. Conversely, overexpression

of MAVS augments interferon prodn. and confers antiviral immunity. Deletion of the CARD-like

domain of MAVS abolishes its signaling function and converts it into a dominant neg. mutant that

inhibits interferon induction. MAVS is cleaved at Cys-508 by the hepatitis C virus NS3/4A protease.

These results reveal a new role of mitochondria in innate immunity, and provide new compn. and

methods for diagnosis and treatment of viral infection and methods of high-throughput screening for

antiviral compds.

75

Page 76: dengue

2007 – 11 patents

1- Kalata B1-based peptides for inhibition of viral NS3 protease. Cui, Taian; Puah, Chum Mok;

Liew, Oi Wah; Lee, Siew Hui. (Singapore Polytechnic, Singapore). PCT Int. Appl. (2007), 56pp.

CODEN: PIXXD2 WO 2007149052 A1 20071227 Designated States W: AE, AG, AL, AM, AT,

AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ,

EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN,

KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, MG, MK, MN, MW, MX, MY, MZ, NA,

NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM,

TN, TR, TT, TZ, UA. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS,

IT, LU, MC, MT, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent

written in English. Application: WO 2007-SG168 20070619. Priority: SG 2006-4180 20060619.

CAN 148:115525 AN 2007:1469276 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Kalata B1-based peptides comprising viral NS3 serine protease recognition sequence on a cyclic mol.

framework for use as anti-virals in the treatment of dengue fever are disclosed. Thus, the peptides

were produced as fusion proteins with His-tagged thioredoxin in transgenic E. coli. After purifn. with

a metal-chelating column the antiviral peptide was released from the fusion protein by enterokinase

cleavage.

2- Inhibition of RNA virus replication using suicide gene- or toxin-encoding viral vector, for use

in prevention or treatment of viral infections in humans. Ventura, Michel; Astier-Gin, Therese;

Litvak, Simon; Dumas, Estelle. (Universite Victor Segalen- Bordeaux 2, Fr.; Centre National Da La

Recherche Scientifique - Cnrs). PCT Int. Appl. (2007), 66pp. CODEN: PIXXD2 WO

2007138193 A1 20071206 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BH,

BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB,

GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK,

LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM,

PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT, TZ.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL,

PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in French.

Application: WO 2007-FR899 20070530. Priority: FR 2006-4806 20060530. CAN 148:45774

AN 2007:1394359 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

76

Page 77: dengue

Abstract

This invention relates to gene therapy for viral infections using a hepatitis C virus (HCV) viral vector

comprising 5'- and 3'-untranslated region (UTR) sequences flanking either a suicide gene or a gene

encoding a toxin, to enable cell death in cells expressing the complete viral replication complex.

Replication of single stranded genomic RNA viruses (such as HCV) requires an intermediate step of

synthesis of the neg. single-strand viral genome, which serves as template for synthesis of the pos.

single-strand virus. For this step of replication, the 5'-UTR and 3'-UTR are indispensable; the 3'-UTR

is responsible for synthesis of the neg. strand from the pos. strand, and the 5'-UTR is responsible for

synthesis of the pos. strand for new virions from the neg. strand template. A genomic fragment contg.

the HCV nonstructural protein genes (NS3-NS5B) was transformed into Huh7 cells. Further a viral

vector was produced that substituted the HCV polyprotein gene (flanked by 5'- and 3'-UTR; the

minimal HCV genomic RNA), with genes encoding the hygromycin resistance protein, FMDV protein

2A, and enhanced green fluorescent protein. The resulting viral vector, 5'-UTR-Hygror-H2AE-EGFP-

3'-UTR, was transformed into the Huh7 cells expressing HCV nonstructural proteins, and viral

replication was demonstrated in hygromycin resistant clones by detection of fluorescence. The viral

vector was then modified to encode toxins or cell death signaling mols., such as ricin A or interferon

regulating factor 1, and cell death was demonstrated in transformed clones. This invention is intended

to be used as pharmaceutical compd. in the prevention or treatment of viral infections from pos.

strand, single-stranded RNA viruses.

3- Antiviral agents that activate RNase L. Silverman, Robert. (The Cleveland Clinic Foundation,

USA). PCT Int. Appl. (2007), 65 pp. CODEN: PIXXD2 WO 2007127212 A2 20071108

Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BH, BR, BW, BY, BZ, CA, CH,

CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR,

HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD,

MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC,

SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT, TZ, UA, UG. Designated States RW: AT, BE,

CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, MT, NL, PT, SE, TR, BF, BJ, CF, CG, CI,

CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2007-US9959

20070425. Priority: US 2006-795069 20060425. CAN 147:496301 AN 2007:1278400 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Activators of RNase L, methods of using the same as antiviral agents, and pharmaceutical compns.

comprising the RNase L activators are disclosed. The activators of RNase L are not cytotoxic, they

77

Page 78: dengue

inhibit smooth muscle cell proliferation in vitro, and therefore have utility in treating restenosis. They

can be also used to treat cancer. Thus, a 2-5A competition binding assay using surface plasmon

resonance on a Biacore model 3000TM was used to det. if an activator interacts with the 2-5A binding

domain of RNase L. The compd. C-5950331 competed with 2-5A for RNase L binding with the

binding const. of 18 μM. The compd. C-5950331 had low cytotoxicity in DU145 and HeLa cells and

showed antiviral activity against vaccinia virus (strain Western Reserve (WR)), a DNA virus in the

pox virus family.

4- Preparation of nitrogen-containing heterocycle derivatives as antiviral agents. Mjalli,

Adnan M.M.; Cooper, Jeremy T.; Arimilli, Murty N.; Andrews, Robert C.; Rothlein, Robert; Altel,

Taleb H. (Transtech Pharma, Inc., USA). U.S. Pat. Appl. Publ. (2007), 53 pp. CODEN:

USXXCO US 2007219239 A1 20070920 Patent written in English. Application: US 2007-

704763 20070209. Priority: US 2006-772309 20060210. CAN 147:385981 AN 2007:1054300

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Title compds. I [R1 = CN, CF3, OCF3, NO2, cycloalkyl, etc.; R2 = halo, NH2, CO2H, OH,

(cyclo)alkyl, (hetero)aryl, etc.; G1 and G2 independently = (un)substituted cycloalkyl, heterocyclyl,

aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalkylheteroaryl, fused

heterocyclylaryl or fused heterocyclylheteroaryl; L1, L2 and L5 independently = direct bond,

(un)substituted alkylene, alkenylene or alkynylene; L3 and L4 independently = direct bond,

(un)substituted alkylene, alkenylene, alkynylene, arylene or heteroarylene; Y1 and Y2 independently =

direct bond, O, C(O), S, OC(O), SO, SO2, etc.; ring A = 5-membered satd. heterocyclyl; V and X

independently = C or N; W, Y or Z independently = O, S, NR5 or CR6; Q = (CR3R4)n, wherein R3-6

independently = H, (un)substituted (cyclo)alkyl, alkylene-cycloalkyl or aryl; CR3R4 = (un)substituted

5- to 7-membered (hetero)cyclyl; n = 0-1; m and p independently = 0-2], and their pharmaceutically

acceptable salts, solvates or prodrugs thereof, are prepd. and disclosed as antiviral agents. Thus, e.g.,

II was prepd. in 11 steps starting from 5-nitroisophthalic acid monomethyl ester and using [(R)-4-

fluorophenethyl]amine. Exemplar compds. of the invention were found to inhibit viral replication in

vaccinia viral assay with an EC50 of ≤ 100 μM, e.g., II showed EC50 value of ≤ 0.5 μM. As antiviral

agents, I should prove useful in the treatment of viral infections and may be administered to a subject

for antiviral therapy or prophylaxis.

5- Anesthetic compound-steroid combination antiviral treatment. Fabunan, Ruben G. (Fil-Am

Tech., Inc., USA). PCT Int. Appl. (2007), 16 pp. CODEN: PIXXD2 WO 2007084548 A2

20070726 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA,

78

Page 79: dengue

CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN,

HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LV, LY,

MA, MD, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS,

RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT, TZ, UA, UG. Designated States

RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF,

CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2007-

US1210 20070117. Priority: US 2006-759847 20060118. CAN 147:181517 AN 2007:817659

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Common colds, influenza, Dengue fever, mumps, measles, hepatitis, rabies, conjunctivitis,

chickenpox, herpes, and HIV infection are treated by i.m. injection of a mixt. comprising a first

ingredient selected from the group consisting of chloroprocaine, tetracaine, chlorotetracaine,

bromoprocaine, proparacaine, fluoroprocaine and benzocaine, and a second ingredient selected from

the group consisting of dexamethasone, flumethasone and betamethasone, or by i.m. injection of a

mixt. comprising a first ingredient selected from the group consisting of procaine, chloroprocaine,

tetracaine, chlorotetracaine, bromoprocaine, proparacaine, fluoroprocaine and benzocaine, and a

second ingredient selected from the group consisting of flumethasone and betamethasone. Compns. of

the invention are administered to prevent HIV from attaching to and penetrating host cells, to penetrate

sanctuary sites to inactivate HIV and to generate vaccines from inactivated HIV.

6- Preparation of antiviral azanucleoside derivatives as inhibitors RNA-dependent viral

polymerases. Chiacchio, Ugo; Mastino, Antonio; Merino, Pedro; Romeo, Giovanni. (Istituto di

Ricerche di Biologia Molecolare P. Angeletti S.p.A., Italy). PCT Int. Appl. (2007), 45pp.

CODEN: PIXXD2 WO 2007065883 A1 20070614 Designated States W: AE, AG, AL, AM, AT,

AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE,

EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR,

KZ, LA, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG, MK, MN, MW, MX, MY, MZ, NA, NG,

NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN,

TR, TT, TZ, UA, UG. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS,

IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent

written in English. Application: WO 2006-EP69288 20061204. Priority: IT 2005-611 20051206.

CAN 147:72983 AN 2007:643846 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

79

Page 80: dengue

Abstract

Azanucleoside analogs I, wherein B is a heterocyclic base, or nucleosidic base; R1 is H, linear or

branched alkyl group, or (un)satd. cycloalkyl groups; R2 is amido, keto or thioketones, linear or

branched alkyl groups, or aryl or benzyl groups; R3 is H or a group convertible in vivo to phosphorous

contg. groups; R4 is H, linear or branched alkyl groups, (un)satd. alkyl groups, or (un)satd. acyl

groups are prepd. and have antiviral activity. Thus, II was prepd. and tested for its HCV-NS3 redn.

assay toxicity (IC50 was 4.8 μM). Further, I can be successfully employed in the treatment of viral

infections such as inhibitor of RNA-dependent RNA polymerases, or viruses from the Flaviviridae,

Orthomyxoviridae, Rheoviridae, Bunyaviridae, or Retroviridase families.

7- Five-membered iminocyclitol derivatives as selective and potent glycosidase inhibitors: new

structures for antivirals and osteoarthritis therapeutics. Liang, Pi-Hui; Lin, Yi-Ling; Wong,

Chi-Huey. (Academia Sinica, Taiwan). PCT Int. Appl. (2007), 33pp. CODEN: PIXXD2 WO

2007067515 A2 20070614 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR,

BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE,

GH, GM, GT, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS,

LT, LU, LV, LY, MA, MD, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH,

PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT, TZ, UA, UG.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT,

SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

Application: WO 2006-US46348 20061205. Priority: US 2005-742406 20051205. CAN

147:46158 AN 2007:642739 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention discloses 5-membered iminocyclitol derivs. which were found to be a potent and

selective inhibitors of the glycoprotein processing α-and β-glucosidase and were further found to be

active antiviral agents against Japanese encephalitis virus, dengue virus serotype 2 (DEN-2), human

SARS coronavirus and human β-hexosaminidase, a new target for development of osteoarthritis

therapeutics. Prepn. of compds. of the invention is included.

8- TMAZ as an antiviral agent and use thereof. Lelas, Tihomir. (Ljubicic, Mijo, Germany;

Ivkovic, Slavko). PCT Int. Appl. (2007), 56pp. CODEN: PIXXD2 WO 2007054085 A2

20070518 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA,

CH, CN, CO, CR, CU, CZ, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR,

HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LV, LY, MA,

MD, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU,

80

Page 81: dengue

SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG,

CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in German. Application: WO 2006-

DE2008 20061110. Priority: DE 2005-102005054306 20051111. CAN 146:514706 AN

2007:538497 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention discloses antiviral pharmaceutical agents comprising esp. tribomechanically activated

zeolites and propolis and/or colostrum. The invention also discloses the use of these agents,

preferably for HIV and hepatitis infections.

9- Pyrazoline derivatives for treatment of viral infections. Ferguson, David M.; Goodell, John.

(Regents of the University of Minnesota, USA). PCT Int. Appl. (2007), 78pp. CODEN: PIXXD2

WO 2007038425 A2 20070405 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG,

BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD,

GE, GH, GM, HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS,

LT, LU, LV, LY, MA, MD, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PG, PH,

PL, PT, RO, RS, RU, SC, SD, SE, SG, SK, SL, SM, SV, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT,

SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

Application: WO 2006-US37241 20060926. Priority: US 2005-721002 20050927. CAN

146:395241 AN 2007:384400 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention provides anti-viral compds. of formula (I) wherein R1-R3 and A have any of the values

defined herein. The invention also provides pharmaceutical compns. comprising such compds. as well

as methods for treating viral infections by administering such compds. to an animal. A prepd. triaryl

pyrazoline deriv. demonstrated significant antiviral activity against West Nile, dengue fever, yellow

fever, and western equine encephalitis viruses.

10- Pyrrolidine derivatives as immunomodulators and antiviral agents. Nash, Robert James;

Carroll, Miles William; Watson, Alison Ann; Fleet, George William John; Horne, Graeme. (MNL

Pharma Limited, UK). PCT Int. Appl. (2007), 72pp. CODEN: PIXXD2 WO 2007010266 A1

20070125 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA,

CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HN, HR,

HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU, LV, LY, MA,

MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS, RU, SC,

81

Page 82: dengue

SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC. Designated States

RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF,

CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2006-

GB2717 20060720. Priority: GB 2005-14865 20050720. CAN 146:177231 AN 2007:88241

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention discloses pyrrolidine compds. and their use in therapy and prophylaxis. In particular,

the invention discloses the use of various pyrrolidine compds. (including N-hydroxyethyl-DMDP and

certain analogs thereof) as immunomodulatory (immunostimulatory or immunosuppressive) drugs

and/or as antivirals (e.g. as glycovirs or alkovirs).

11- Preparation of 2,3,4,9-tetrahydro-1H-β -carbolines for treatment of dengue fever, yellow

fever, West Nile virus, and hepatitis C virus infection. Gudmundsson, Kristjan. (Smithkline

Beecham Corporation, USA). PCT Int. Appl. (2007), 53pp. CODEN: PIXXD2 WO

2007002051 A1 20070104 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR,

BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE,

GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LA, LC, LK, LR, LS, LT, LU,

LV, LY, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO,

RS, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT,

SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

Application: WO 2006-US23928 20060619. Priority: US 2005-692810 20050622. CAN

146:100663 AN 2007:13571 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Title compds. I [wherein each R1 = halo, (halo)alkyl, alkenyl, etc.; p = 0-4; n = 0 or 1; X = C(O),

C(O)O, SO2, etc.; R2, R3 = (un)substituted (hetero)aryl or heterocyclyl] and pharmaceutically

acceptable salts, solvates, and physiol. functional derivs. thereof were prepd. for the treatment of

infection due to Flaviviridae, such as flaviviruses, pestiviruses, and hepaciviruses, particularly dengue

fever, yellow fever, West Nile virus, or HCV. Thus, cyclization of 5-bromotryptamine hydrochloride

with p-tolualdehyde in glacial acetic acid (80%) followed by treatment with benzyl chloroformate led

to II. This compd. showed anti-HCV activity with IC50 = 5 nM.

82

Page 83: dengue

2006 – 14 patents

1- Preparation of nucleosides with non-natural bases as anti-viral agents. Storer, Richard;

Gosselin, Gilles; Griffon, Jean-Francois; Pierra, Claire. (Idenix (Cayman) Limited, Cayman I.; Centre

Nationale De La Recherche Scientifique). Can. Pat. Appl. (2006), 149pp. CODEN: CPXXEB

CA 2600359 A1 20060909 Patent written in English. Application: CA 2006-2600359 20060309.

Priority: US 2005-660117 20050309; WO 2006-IB2550 20060309. CAN 148:79268 AN

2007:1439303 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The prepn. nucleosides with non-natural bases I, wherein R1 is H or a halogen; R2 is an H or halogen,

but preferably F; R3 is (un)substituted amino; Q is independently N, CH or NH; and Z can be a

(un)substituted ribofuranosyl moiety, is presented. Further, Z can be modified as II, such that the Base

is described as I above; X is O, S, or NH; R is independently H, a substituted phosphate, phosphonate,

acyl, alkyl or amino acid group; A can be independently H, hydroxy, alkyl, alkenyl, halo, etc.; B is

independently H, alkyl, a halo substituted alkyl or a halogen. Thus, III was prepd. and tested treating a

host infected with flavivirus, pestivirus or hepacivirus (EC50 less than 15 μM). Subsequently, β-D

and β-L nucleosides of I are to be used as therapeutic prodrugs treating diseases related to viral

origins.

2- Acylguanidines as antiviral compounds and their preparation, pharmaceutical compositions

and use in the treatment of viral infections in mammals. Ewart, Gary Dinneen; Best, Wayne

Morris. (Biotron Limited, Australia). PCT Int. Appl. (2006), 49pp. CODEN: PIXXD2 WO

2006135978 A1 20061228 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR,

BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE,

GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV,

LY, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RS,

RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT,

SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

Application: WO 2006-AU880 20060623. Priority: AU 2005-903360 20050624. CAN 146:100349

AN 2006:1356781 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention relates to acylguanidine compds. of formula I and compns. having antiviral

activity. Compds. of formula I wherein R1 is (un)substituted (un)fused cinnamyl, (un)substituted

naphthyl, and (un)substituted phenyl; and their pharmaceutically acceptable salts thereof, are claimed.

The invention also relates to methods for the therapeutic or prophylactic treatment of viral infections

83

Page 84: dengue

in mammals. Example compd. II was prepd. by olefination of 2,3-methylenedioxybenzaldehyde with

tri-Et phosphonoacetate; the resulting Et 2,3-methylenedioxycinnamate underwent hydrolysis to give

the corresponding cinnamic acid, which underwent amidation with guanidine to give compd. II. All

the invention compds. were evaluated for their antiviral activity, toxicity (TC50)and calcn. of the

antiviral index (AI). From the assay it was detd. that compd. II exhibited an IC50 value 1.1 μM, TC50

of > 100 μM, and AI > 100.

3- Preparation and antiviral activity of D-glucopyranose 1-[3,5-bis(1,1-dimethylethyl)-4-

hydroxybenzoate]. Vachy, Robert. (RDW Pharma, Fr.). Fr. Demande (2006), 16pp.

CODEN: FRXXBL FR 2887249 A1 20061222 Patent written in French. Application: FR 2005-

6304 20050621. Priority: . CAN 146:39014 AN 2006:1338924 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

FR 2887249 A1 20061222 FR 2005-6304 20050621

FR 2887249 B1 20070928

Priority Application

FR 2005-6304 20050621

Abstract

D-Glucopyranose 1-[3,5-bis(1,1-dimethylethyl)-4-hydroxybenzoate] was prepd. by esterification of D-

glucose with 3,5-di-tert-butyl-4-hydroxybenzoic acid and used as antiviral agent for treatment of

Herpes, AIDS, influenza of hepatitis B and C, dengue, and Ebola viruses. Title compd. was tested in

vitro as antiviral agent and showed better soly. of BHT and 3,5-di-tert-butyl-4-hydroxybenzoic acid.

4- Antisense antiviral oligonucletides targeting RNA stem-loop structure for treating infections

of ssRNA viruses. Iversen, Patrick L.; Stein, David A.; Weller, Dwight D. (Avi Biopharma, Inc.,

USA). U.S. Pat. Appl. Publ. (2006), 64pp., Cont.-in-part of U.S. Ser. No. 226,995. CODEN:

USXXCO US 2006269911 A1 20061130 Patent written in English. Application: US 2006-

432031 20060510. Priority: US 2004-611063 20040916; US 2005-226995 20050914. CAN

146:20246 AN 2006:1256345 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention provides antisense antiviral compds. and methods of their use and prodn. in inhibition

of growth of viruses of the Flaviviridae, Picomoviridae, Caliciviridae, Togaviridae, Arteriviridae,

84

Page 85: dengue

Coronaviridae, Astroviridae and Hepeviridae families in the treatment of a viral infection. The

antisense antiviral compds. are substantially uncharged morpholino oligonucleotides having a

sequence of 12-40 subunits, including at least 12 subunits having a targeting sequence that is

complementary to a region assocd. with stem-loop secondary structure within the 5'-terminal end 40

bases of the pos.-sense RNA strand of the virus. The antisense compds. are preferably

phosphorodiamidate-linked morpholino oligonucleotide (PMO) analogs. These PMOs may be

conjugates to Arg-rich peptides to enhance their cellular uptake. Thus, such antisense PMOs were

prepd. and shown to inhibit yellow fever virus, dengue virus, porcine reproductive and respiratory

syndrome virus, tick-borne encephalitis virus, and west nile virus in in vitro assays.

5- Preparation of carbazoles and related compounds for treatment of dengue fever, yellow fever,

west nile virus, and hepatitis C virus infection. Gudmundsson, Kristjan. (Smithkline Beecham

Corporation, USA). PCT Int. Appl. (2006), 59pp. CODEN: PIXXD2 WO 2006121467 A2

20061116 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA,

CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU,

ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG,

MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK,

SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG,

CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2005-

US41091 20051114. Priority: US 2004-630166 20041122. CAN 145:489114 AN 2006:1207231

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Title compds. [I; n = 0-2; X = NH, O, S, SO, SO2; R, R1 = halo, haloalkyl, alkyl, alkenyl, alkynyl,

cycloalkyl, aryl, aryloxy, arylamino, heterocyclyl, heterocyclyloxy, heterocyclylamino, cyano, NO2,

N3, etc.; p, q = 0-5; A = aryl, heteroaryl], were prepd. for the treatment of infection due to flaviviruses,

pestiviruses, and hepaciviruses. Thus, 6-chloro-N-phenyl-2,3,4,9-tetrahydro-1H-carbazol-1-amine

(prepn. outlined) showed anti-HCV activity with IC50 = 5 nM.

6- Tetrahydrocarbazoles useful as inhibitors of hepatitis C and other viruses belonging to

Flaviviridae. Gudmundsson, Kristjan; Samano, Vicente. (Smithkline Beecham Corporation,

USA). PCT Int. Appl. (2006), 69pp. CODEN: PIXXD2 WO 2006118607 A2 20061109

Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN,

CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL,

IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG, MK,

85

Page 86: dengue

MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL,

SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA. Designated States RW: AT,

BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI,

CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2005-US41089

20051114. Priority: US 2004-629905 20041122. CAN 145:465666 AN 2006:1179153 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention relates to tetrahydrocarbazoles [shown as I; n = 0-2; t = 0 or 1; X = NH, O,

-R10-, -OR10-, -R10O-, -R10OR10-, NR10-, -R10N-, -R10NR10-, -R10S(O)m-, or R10S(O)mR10-;

Y = C(O), S(O)m; R and R1 independently = halo, haloalkyl, alk(en/yn)yl, cycloalkyl, cyano, nitro or

azido, et al.; m = 0-2; R10 = alkylene, cycloalkylene, alkenylene, cycloalkenylene, and alkynylene; p,

q = 0-5; A = (hetero)aryl; addnl. details including provisos are given in the claims; or salts, solvates

and physiol. functional derivs. thereof] that are useful in the treatment of viruses belonging to

Flaviviridae, including flaviviruses, pestiviruses, and hepaciviruses. The invention includes compds.

useful for the treatment or prophylaxis of dengue fever, yellow fever, West Nile virus, and HCV.

Although the methods of prepn. are not claimed, prepns. and/or characterization data for .apprx.70

examples of I are included. For example, 6-chloro-2,3,4,9-tetrahydro-1H-carbazol-1-amine was

reacted with picolinoyl chloride to give II as a racemate in 63% yield, which was then sepd. into two

pure enantiomers by supercrit. fluid chromatog. IC50 values for inhibition of HCV activity are

tabulated for 6 examples of I, e.g. 6 nM for (R)-II.

7-Antiviral artificial CpG-containing single-stranded oligodeoxynucleotides in combination with

ribavirin. Wang, Li-Ying; Bao, Mu-Sheng; Yu, Yong-Li. (Changchun Huapu Biotechnology Co.,

Ltd., Peop. Rep. China). PCT Int. Appl. (2006), 74pp. CODEN: PIXXD2 WO 2006108358 A1

20061019 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA,

CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU,

ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG,

MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK,

SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG,

CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in Chinese. Application: WO 2006-CN667

20060413. Priority: CN 2005-10064537 20050413. CAN 145:443761 AN 2006:1093278

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

86

Page 87: dengue

Abstract

The present invention provides a compn. contg. ribavirin and artificial CpG single-stranded

oligodeoxynucleotides or a kit which can prevent or treat viral infection and infectious diseases caused

by virus, and use thereof. The invention also provides a method for preventing or treating viral

infection and infectious diseases caused by virus.

8- Sulfur oligonucleotides having antiviral activities. Vaillant, Andrew; Juteau, Jean-Marc.

(Replicor Inc., Can.). PCT Int. Appl. (2006), 51pp. CODEN: PIXXD2 WO 2006096995 A1

20060921 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA,

CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU,

ID, IL, IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG,

MK, MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK,

SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG,

CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2006-CA411

20060317. Priority: US 2005-662364 20050317. CAN 145:348566 AN 2006:978227 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Sulfur oligonucleotides having antiviral activities through a sequence-independent mode of action are

described, along with their use as broad spectrum therapeutic agents for treating viral infections. The

sulfur modification may be at any location (e.g., on the base moiety, sugar moiety, or internucleotidic

linkages) in order to confer the necessary chem. properties required for the sequence-independent

antiviral activity of oligonucleotides. The sulfur modification is necessary and can be addnl. to

another modification, like a phosphorothioate or phosphorodithioate modification. It is not necessary

for the oligonucleotide to be complementary to any viral sequence or to have a sequence-dependent

aptameric activity or to have a particular distribution of nucleotides in order to have activity. Different

lengths of random sequence sulfur-modified oligonucleotides have varying effect, with an optimal

length of 30-40 bases. Also described are methods for treatment of viral infections, cancers caused by

oncogene viruses, and other diseases whose etiol. is viral-based. Such method of treatment comprises

the administration of sulfur oligonucleotides to a human or an animal in an acceptable form alone or in

combination with another antiviral compd.

9- Preparation of quinazoline derivatives as antiviral agents. Cockerill, George Stuart; Flack,

Stephen, Sean; Mathews, Neil; Salter, James Iain. (Arrow Therapeutics Limited, UK). PCT Int.

Appl. (2006), 27pp. CODEN: PIXXD2 WO 2006079833 A1 20060803 Designated States W:

87

Page 88: dengue

AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE,

DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM,

KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG, MK, MN, MW, MX, MZ, NA,

NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR,

TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA. Designated States RW: AT, BE, CH, CY, DE, DK, ES,

FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE,

SN, TD, TG. Patent written in English. Application: WO 2006-GB294 20060130. Priority: GB

2005-1964 20050131; US 2005-649564 20050204; US 2005-668456 20050405. CAN 145:211057

AN 2006:765057 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Quinazoline derivs. [I; X = direct bond, LNR; R = H, C1-C4 alkyl; L = C1-C4 alkylene, C6-C10

arylene, or 5-to-10-membered heteroarylene; R1 = H, C6-C10 aryl, C1-C4 (hydroxy)alkyl; R2 = C6-

C10 aryl, C1-C4 (hydroxy)alkyl; R3 = C6-C10 (un)substituted aryl, C3-C6 (un)substituted

carbocyclyl, 5-to-10-membered (un)substituted heteroaryl, 5-to-10-membered (un)substituted

heterocyclyl; NR1R2 = 5-to-10-membered heterocyclyl, 5-to-10-membered heteroaryl; e.g., N-(4-

morpholinophenyl)-6-[4-(4-morpholinophenylamino)quinazolin-6-yl]quinazolin-4-amine], which are

active in inhibiting the replication of flaviviridae, are prepd.

10- Antiviral activity from medicinal mushrooms. Stamets, Paul. (USA). U.S. Pat. Appl. Publ.

(2006), 14pp., Cont.-in-part of U.S. Ser. No. 145,679. CODEN: USXXCO US 2006171958 A1

20060803 Patent written in English. Application: US 2006-386402 20060322. Priority: US 2004-

534776 20040106; US 2005-29861 20050104; US 2005-145679 20050606. CAN 145:195587

AN 2006:763418 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Compds. having unique antiviral properties are prepd. from medicinal mushroom mycelium, exts. and

derivs. The compns. are derived from Fomitopsis, Piptoporus, Ganoderma and blends of medicinal

mushroom species and are useful in preventing and treating viruses including Orthopox viruses,

influenza, avian influenza, Venezuelan Equine Encephalitis, yellow fever, West Nile, Dengue, New

World and Old World arenaviruses, hantavirus, Rift Valley fever, sandfly fever, hantavirus, SARS,

Rhinovirus and other viruses.

11- Antiviral drug combinations using a glycosylation modulator and a membrane fusion

inhibitor. Nash, Robert James; Slingsby, Jason H.; Carroll, Miles William. (MNL Pharma Limited,

UK). PCT Int. Appl. (2006), 85 pp. CODEN: PIXXD2 WO 2006077427 A2 20060727

Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN,

88

Page 89: dengue

CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL,

IN, IS, JP, KE, KG, KM, KN, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, LY, MA, MD, MG, MK,

MN, MW, MX, MZ, NA, NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL,

SM, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA. Designated States RW: AT,

BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI,

CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2006-GB209

20060120. Priority: GB 2005-1352 20050121. CAN 145:180926 AN 2006:733122 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention discloses a combined prepn. comprising a glycosylation modulator and a membrane

fusion inhibitor for combined, simultaneous, or sequential use in the treatment of infections caused by

viruses bearing glycosylated envelope proteins.

12- Antiviral antisense oligonucleotides for treating ss(+)RNA viral infection. Iversen, Patrick

L.; Stein, David A. (USA). U.S. Pat. Appl. Publ. (2006), 62 pp. CODEN: USXXCO US

2006063150 A1 20060323 Patent written in English. Application: US 2005-226995 20050914.

Priority: US 2004-611063 20040916. CAN 144:306424 AN 2006:273262 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Abstract

The invention provides antisense antiviral compds. for inhibition of replication of Flaviviridae,

Picornoviridae, Caliciviridae, Togaviridae, Arteriviridae, Coronaviridae, Astroviridae and Hepeviridae.

The antisense antiviral compds. are substantially uncharged morpholino oligonucleotides having a

sequence of 12-40 subunits, including at least 12 subunits having a targeting sequence that is

complementary to a region assocd. with stem-loop secondary structure within the 5'-terminal end 40

bases of the pos.-sense RNA strand of the virus. The antisense oligonucleotides may be conjugated to

an arginine-rich peptide to promoter uptake into infected host cells. Thus, a phosphorodiamidate-

linked morpholino oligonucleotide targeting the 5'-terminal of West Nile virus RNA conjugated to

(RAhxR)4AhxβAla (Ahx = 6-aminohexanoic acid; βAla = β-alanine) was shown to prolong survival

of mice infected with the virus.

13- Preparation of nucleoside-lipid conjugates as antiviral and antitumor agents. Ahmad,

Moghis U.; Ali, Shoukath M.; Khan, Abdul R.; Ahmad, Imran. (Neopharm, Inc., USA). PCT Int.

Appl. (2006), 72 pp. CODEN: PIXXD2 WO 2006029081 A2 20060316 Designated States W:

AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE,

DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM,

89

Page 90: dengue

KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI,

NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ,

UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE, DK, ES,

FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE,

SN, TD, TG. Patent written in English. Application: WO 2005-US31543 20050902. Priority: US

2004-606610 20040902. CAN 144:292980 AN 2006:235096 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Abstract

The invention provides methods for synthesizing nucleoside-lipid conjugates I, wherein Y1 and Y2 are

the same or different and are -O-C(O)-, -O-, -S-, -NH-C(O)- or the like; R1 and R2 are independently

H, satd. alkyl group and unsatd. alkyl group; X is H, alkyl group and a cation; R3 is a nucleoside

selected from a group consisting of cytosine, guanine, adenine, thymine, uracil, inosine, xanthine and

hypoxanthine; R4 and R5 are independently hydrogen, hydroxy, halo group, nitro, alkyl group,

substituted alkyl and alkoxy group; R6 is hydrogen, hydroxy group, azido group, amino group, alkyl

group, halo group and substituted amino; five membered cyclic sugar is selected from a group

consisting of ribofuranose, arabinofuranose, deoxyribofuranose and xylofuranose having varying fatty

acid and alkyl chain lengths with or without unsatn. and their use in the treatment of cancer and viral

diseases. More particularly, the invention provides methods for prepg. gemcitabine-cardiolipin

conjugates, and analogs thereof, cytarabine-cardiolipin conjugates, and analogs thereof. Addnl., the

methods of the invention comprise administering a compd. of invention as prodrug or a

pharmaceutical prepn. to combat mammalian diseases, preferably cancer, viral infections and bone

disorders. The cancer is selected from a group consisting of cancers of the head, neck, brain, blood,

breast, lung, pancreas, bone, spleen, bladder, prostate, testes, colon, kidney ovary, and skin. The viral

disease is selected from a group consisting of HIV, Herpes simplex viruses, human Herpes virus 6,

human Herpes virus 7, human Herpes virus 8, Ebola virus, Influenza virus, Tuberculosis, Hepatitis A,

Hepatitis B, Hepatitis C, Hepatitis D, Hepatitis E, Parainfluenza virus, Respiratory syncytial virus,

Cholera, pneumonia, SARS virus, West Nile virus, Respiratory syncytial virus, Dengue virus, Corona

viruses, Vaccinia virus, Cytomegalovirus, human Rhinovirus, Papilloma virus, and Human

Herpesvirus 4.

The bone disorder is selected from a group consisting of osteoporosis, Paget's disease, metastatic bone

cancers, hyperparathyroidism, rheumatoid arthritis, Gaucher's disease. Thus, 5'-O-succinyl[2-O-1,3-

bis(1,2-O-dimyristoyl-sn-glycero)-3-phosphorylglycerol dimethylester] gemcitabine was prepd. and

tested in-vitro and in mice as antiviral and antitumor agent. The toxicity of gemcitabine-cardiolipin

conjugate at 18 μmol/kg after 6 daily treatments and the body wt. loss on day 7 was significantly less

90

Page 91: dengue

compared to gemcitabine. When mice were treated with gemcitabine-cardiolipin conjugate at 18

μmol/kg for 5 days, the max. body wt. loss was only 3 % compare to 22 % for gemcitabine.

14- Improved synthesis of sodium pentaborate pentahydrate (NaB5O8.5H2O) for use in

antibacterial and antiviral compositions. Galvan Perez, Juan Pablo. (Mex.). PCT Int. Appl.

(2006), 81 pp. CODEN: PIXXD2 WO 2006025724 A1 20060309 Designated States W: AE,

AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK,

DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KP,

KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI, NO,

NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA,

UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI,

FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN,

TD, TG. Patent written in Spanish. Application: WO 2005-MX79 20050902. Priority: MX 2004-

8485 20040902. CAN 144:265950 AN 2006:213041 CAPLUS (Copyright (C) 2010 ACS on

SciFinder (R))

Abstract

The invention relates to an improved method for the synthesis of sodium pentaborate pentahydrate

(NaB5O8.5H2O), which consists in reacting disodium tetraborate decahydrate (borax) with orthoboric

acid (boric acid). The method is performed over a period of no more than five days and does not

require the use of fungal strains. Pharmaceutical compns. contg. NaB5O8.5H2O are used to boost the

immune system against ailments caused by bacteria and viruses. Thus, NaB5O8.5H2O was effective

against 14 bacterial species in vitro at 400 mg/mL.

91

Page 92: dengue

2005 – 14 patents

1- Potent and selective inhibition of viral replication by aurintricarboxylic acid. He, Runtao;

Andonov, Anton; Cao, Jingxin; Drebot, Mike; Li, Xueguang. (Canada, Minister of Health, Can.).

PCT Int. Appl. (2005), 64 pp. CODEN: PIXXD2 WO 2005123965 A1 20051229 Designated

States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU,

CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE,

KG, KM, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA,

NG, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR,

TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE,

DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML,

MR, NE, SN, TD, TG. Patent written in English. Application: WO 2005-CA924 20050615.

Priority: US 2004-579247 20040615; US 2004-608862 20040913. CAN 144:81142 AN

2005:1354624 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Described herein is a method of inhibiting replication of an organism which has an essential enzyme

which includes a binding groove that is bound by ATA comprising administering to a patient in need of

such treatment an effective amt. of aurintricarboxylic acid (ATA). In this study, the inventors analyzed

the effect of aurintricarboxylic acid (ATA) on SARS-CoV replication in cell culture, and found that

ATA could drastically inhibit SARS-CoV replication, with viral prodn. being more than 1000 fold than

that in the untreated control. As can be seen in Figure 4, a putative 3D structure of SARS RNAP and

ATA (using an ATA ligand model) shows the template binding groove of RNAP which is also bound

by ATA. That is, ATA binding at this groove prevents template binding and therefore viral replication

by RNAP. Furthermore, as can be seen from the sequence comparison shown in Figure 5, enzymes

from other organisms known to be inhibited by ATA have similar grooves. In addn., the inhibitory

effect of aurintricarboxylic acid (ATA) on vaccinia virus replication in tissue culture is described

herein. Concns. of ATA in the range of 400 μg/mL decreased viral replication as much as 250,000 fold

as compared to controls. A block in replication was evident at drug concns. as low as 25 pg/mL.

Inhibition of the viral phosphatase, H1L, which is essential for virus replication, was found to be one

mechanism through which ATA exerts its antiviral effect. Western blotting also revealed that the ERK

signaling cascade was down-regulated in cells treated with ATA. The activity of the ERK signaling

cascade has previously been implicated in the vaccinia virus lifecycle. As discussed below, ATA is an

effective antiviral for coronaviruses, for example, SARS-CoV, West Nile Virus, Norwalk, Dengue and

Japanese Encephalitis virus.

92

Page 93: dengue

2- Interferon combination therapy. Seiwert, Scott D.; Blatt, Lawrence M.; Tan, Hua; Derrick,

Jena Rey; Hong, Jin; Radhakrishnan, Ramachandran. (Intermune, Inc., USA). PCT Int. Appl.

(2005), 134 pp. CODEN: PIXXD2 WO 2005123113 A2 20051229 Designated States W: AE,

AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK,

DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KP,

KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NG, NI, NO,

NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN, TR, TT, TZ, UA,

UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI,

FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN,

TD, TG. Patent written in English. Application: WO 2005-US21050 20050614. Priority: US

2004-579625 20040614. CAN 144:86581 AN 2005:1350327 CAPLUS (Copyright (C) 2010

ACS on SciFinder (R))

Abstract

The authors disclose combination therapy comprising interferon-α (IFN-α) and IFN-γ. The

combination therapy is useful for treating viral infections, fibrotic disorders, and proliferative diseases.

3- Preparation of morpholinylanilino quinazoline derivatives for use as antiviral agents.

Spencer, Keith; Dennison, Helena; Matthews, Neil; Barnes, Michael; Chana, Surinder. (Arrow

Therapeutics Limited, UK). PCT Int. Appl. (2005), 55 pp. CODEN: PIXXD2 WO 2005105761

A1 20051110 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ,

CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR,

HU, ID, IL, IN, IS, JP, KE, KG, KM, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK,

MN, MW, MX, MZ, NA, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM,

SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW:

AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG,

CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English. Application: WO 2005-

GB1598 20050428. Priority: GB 2004-9494 20040428; GB 2004-25268 20041116. CAN

143:460186 AN 2005:1193587 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Title compds. I [wherein R1 - R4 = H, alkyl, haloalkyl, etc., and pharmaceutically acceptable salts

thereof] were prepd. as antiviral agents. For instance, thermal cyclization of 5-bromo-2-aminobenzoic

acid with formamide followed by chlorination with thionyl chloride gave crude 6-bromo-4-

chloroquinazoline, which was condensed with 4-morpholinoaniline in refluxing acetonitrile to afford

II. This compd. showed activity in reducing the replicon level with IC50 of < 5 μM and in reducing

93

Page 94: dengue

the cell area with TD50 of >25 μM in the cell culture assay using HCV replicon cells Huh 9B.

Therefore, I and their pharmaceutical compns. are effective in treating or preventing flaviviridae

infections.

4- Preparation of novel sugar chain-supported carbosilane dendrimer. Terunuma, Daiyo;

Hatano, Ken; Suzuki, Yasuo; Jwa, Ilpall. (Saitama University, Japan; Shizuoka Prefecture; Matsuoka,

Koji). PCT Int. Appl. (2005), 57 pp. CODEN: PIXXD2 WO 2005103064 A1 20051103

Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN,

CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL,

IN, IS, KE, KG, KM, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX,

MZ, NA, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SM, SY, TJ, TM, TN,

TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY,

DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA,

ML, MR, NE, SN, TD, TG. Patent written in Japanese. Application: WO 2005-JP7755 20050419.

Priority: JP 2004-124757 20040420. CAN 143:440686 AN 2005:1171104 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Abstract

Sugar chain (oligosaccharide)-supported-carbosilane dendrimers represented by the following general

formula (R1)mSi{-R2-Si(R6)l[R3-Si(R7)k(R4-S-R5-A)3-k]3-l}n, the following general formula

(R1)mSi[-R2-Si(R6)l(R4-S-R5-A)3-l], or the following general formula (R1)mSi(R4-S-R5-A)n (R1,

R6, R7 = C1-6 alkyl, Ph, vinyl, allyl; R2, R3, R4, R5 = C1-6 alkylene, phenylene, alkenylene; A =

paraglobosyl (Q) (R = H), α-mannopyranosyl, group derived by removing H from 1-OH group of

mannooligosaccharide contg. 2-8 mannose; m = an integer of 0-3, n = an integer of 1-4, and m+n=4; k,

l = 1, 0-2) are prepd. A Dengue fever virus infection inhibitor, an antiviral agent, and a target

substance for the screening of anti-HIV agents contg. the sugar chain-supported-carbosilane dendrimer

as an active ingredient are also disclosed. Thus, 27.9 mg carbosilane dendrimer of formula

Me2Si[(CH2)3Si(CH2CH2Br)3]2 and 345.3 mg paragloboside Q(CH2)5SAc (R = Ac) were dissolved

in 0.4 mL dry DMF, treated with dry MeOH, thoroughly stirred, treated dropwise with 0.280 mL 1 M

NaOMe/MeOH, stirred at room temp. for 11 h, neutralized by adding 0.2 mL AcOH, concd., treated

with dry pyridine and 2.5 mL Ac2O, stirred at 30ϒ for 4 h to give 44% (110.0 mg)

[[Q(CH2)5S(CH2)3]3SiCH2CH2]2SiMe2 (R = Ac) (I). I (69.6 mg) was dissolved in 1.0 mL dry

MeOH, treated with 3.5 mg NaOMe, stirred at room temp. for 30 min, treated with 5.0 mL 0.1M aq.

NaOH soln., stirred for 5 h, further treated with 5.0 mL 0.1M aq. NaOH soln., stirred for 21 h,

neutralized with Amberlite IR120B, concd., and purified by gel permeation chromatog. (GPC) to give

[[Q(CH2)5S(CH2)3]3SiCH2CH2]2SiMe2 (R = H) (II). II in vitro lowered the infection rate of

94

Page 95: dengue

BHK21 cells by Dengue fever virus to 47.16 and 53.95% at 0.5 and 1 mM, resp., from 100% for

control cells vs. 2.26% for heparin 100 μg/mL.

5- Method for discovery and development of broad-spectrum antiviral drugs. Buscher,

Benjamin A.; Dyall, Julie; Jockel-Balsarotti, Jennifer I.; O'Guin, Andrew K.; Olivo, Paul D.; Roth,

Robert M.; Zhou, Yi. (USA). U.S. Pat. Appl. Publ. (2005), 36 pp. CODEN: USXXCO US

2005164167 A1 20050728 Patent written in English. Application: US 2004-766226 20040128.

Priority: . CAN 143:146652 AN 2005:672730 CAPLUS (Copyright (C) 2010 ACS on SciFinder

(R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20050164167 A1 20050728 US 2004-766226 20040128

Priority Application

US 2004-766226 20040128

Abstract

The invention discloses a method for identifying a broad-spectrum antiviral lead compd. Also

disclosed are methods for marketing and delivering a broad-spectrum antiviral compd. and methods

for treating patients with antiviral infections with a broad-spectrum antiviral drug.

6- Method of regulating phosphorylation of sr protein and antiviral agents comprising sr protein

activity regulator as the active ingredient. Hagiwara, Masatoshi; Fukuhara, Takeshi; Suzuki,

Masaaki; Hosoya, Takamitsu. (Japan). PCT Int. Appl. (2005), 122 pp. CODEN: PIXXD2 WO

2005063293 A1 20050714 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR,

BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE,

GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD,

MG, MK, MN, MW, MX, MZ, NA, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK,

SL, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States

RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IS, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF,

CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in Japanese. Application: WO 2004-

JP19393 20041224. Priority: JP 2003-435085 20031226. CAN 143:126753 AN 2005:612118

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

It is intended to provide: (1) antiviral agents lowering or inhibiting the activity of an SR protein, more

specifically speaking, (i) an antiviral agent promoting the dephosphorylation of an SR protein and (ii)

95

Page 96: dengue

an antiviral agent inhibiting a protein phosphorylating an SR protein; (2) an antiviral agent inhibiting

the expression of an SR protein; and (3) an antiviral agent activating a protein having an opposite

function to an SR protein. It is also intended to provide compds. which inhibit SRPK phosphorylating

an SR protein. These compds. inhibit the activity of the SR protein and show an antiviral effect.

Thus, antiviral agents which are efficacious against a novel virus and widely applicable and show a

highly sustained effect are provided to cope with the occurrence of various novel viruses.

7- A West Nile virus (WNV) reverse genetics dual-reporter system for high throughput cell-based

screening and identifying antivirals and vaccines against flaviviral infections. Shi, Pei-Yong;

Lo, Michael; Tilgner, Mark. (Health Research, Inc., USA). U.S. Pat. Appl. Publ. (2005), 81 pp.

CODEN: USXXCO US 2005058987 A1 20050317 Patent written in English. Application: US

2003-706892 20031113. Priority: US 2002-427117 20021118. CAN 142:309857 AN

2005:238534 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Patent Family Information

Patent No. Kind Date Application No. Date

US 20050058987 A1 20050317 US 2003-706892 20031113

US 7355033 B2 20080408

Priority Application

US 2002-427117P P 20021118

Abstract

The invention relates to compns. and methods for the identifying novel chemotherapeutics and

vaccines effective against flaviviral infections, such as, West Nile virus (WNV) and other emerging

flaviviruses, such as, Japanese encephalitis virus (JEV), St. Louis encephalitis virus (SLEV),

Alkhurma virus (AV), Kadam virus (KV), Jugra virus (JV), Cacipacore virus (CV), Yaounde virus

(YV), Tick-borne encephalitis virus (TBEV), Dengue viruses (DENV-1, DENV-2, DENV-3, DENV-

4), Yellow fever virus (YFV) and Murray Valley encephalitis virus (MVEV). The instant invention

provides stable and novel lineage I WNV reverse genetics systems, and methods for making the

reverse genetics systems , specifically, a fully-infectious WNV cDNA or replicon system engineered

with one or more nucleotide sequences each encoding a reporter gene to be used in high throughput

cell-based screening assays for the identification of antiflaviviral chemotherapeutics and/or vaccines

effective to treat and/or immunize against infections by WNV and other flaviviruses. The present

invention further provides methods of high throughput screening of antiflaviviral compds. or improved

derivs. thereof using novel lineage I WNV reverse genetics systems and/or cell lines stably contg. the

96

Page 97: dengue

reverse genetics systems. Also, the invention provides novel pharmaceutical compns. comprising an

attenuated lineage I WNV that is less virulent but similarly immunogenic as the parent WNV and is

capable of providing a protective immune response in a host.

8- Antimetabolite antiviral dosing regimen for hepatitis C virus or flaviviridae therapy.

Stuyver, Lieven J. (Belg.). U.S. Pat. Appl. Publ. (2005), 23 pp. CODEN: USXXCO US

2005049220 A1 20050303 Patent written in English. Application: US 2004-921052 20040818.

Priority: US 2003-496202 20030818. CAN 142:254563 AN 2005:185375 CAPLUS (Copyright

(C) 2010 ACS on SciFinder (R))

Abstract

An anti-hepatitis C agent which is an antimetabolite to the host and cannot be administered on a daily

or chronic basis as is usual in antiviral therapy (referred to below as an "anti-HCV antimetabolite"),

can be administered using a traditional anticancer dosing regimen (for example via i.v. or parenteral

injection), over a period of 1-7 days followed by cessation of therapy until rebound of the viral load is

noted. This dosing regimen runs counter to conventional antiviral experience, wherein effective

agents are usually administered over at least fourteen days of sustained therapy, and typically on an

indefinite daily basis.

9- Virally encoded RNAs as substrates, inhibitors, and delivery vehicles for RNAi and uses for

antiviral therapy. Kowalik, Timothy F.; Stadler, Bradford M. (University of Massachusetts,

USA). PCT Int. Appl. (2005), 123 pp. CODEN: PIXXD2 WO 2005019433 A2 20050303

Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN,

CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL,

IN, IS, JP, KE, KG, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX,

MZ, NA, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR,

TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE,

DK, ES, FI, FR, GB, GR, IE, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR,

NE, SN, TD, TG. Patent written in English. Application: WO 2004-US27436 20040823. Priority:

US 2003-497156 20030822; US 2004-566114 20040427. CAN 142:273968 AN 2005:182814

CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention provides methods for identifying druggable targets in assays that feature

compns., cells and/or organisms having structured viral non-coding RNAs (svRNAs) and an RNA

interference (RNAi) pathway. Methods for identifying antiviral agents and creating vaccines are also

featured. The invention further provides methods for inhibiting RNAi involving svRNAs or inhibitory

97

Page 98: dengue

derivs. thereof. The invention also provides compns. for delivering siRNA and miRNA mols. derived

from svRNA loci and methods of use thereof. Antiviral vaccine comprising vectors encoding siRNA

or miRNA from VA RNA locus are also featured.

10- The artificial CpG single strand deoxidation oligonucleotide and its antiviral uses. Yu,

Yongli; Wang, Liying. (Changchun Huapu Biotechnology Co., Ltd., Peop. Rep. China). PCT Int.

Appl. (2005), 22 pp. CODEN: PIXXD2 WO 2005014611 A1 20050217 Designated States W:

AE, AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE,

DK, DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP,

KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NI, NO, NZ,

OM, PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US,

UZ, VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB,

GR, IE, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG.

Patent written in Chinese. Application: WO 2004-CN863 20040726. Priority: CN 2003-146157

20030725; CN 2003-156224 20030905. CAN 142:212308 AN 2005:141083 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The present invention disclosed a set of artificial ODNs which contain one or more CpG(s), the said

ODNs can stimulate human PBMC to produce antiviral substances. These antiviral substances make

cell avoid of the attack from virus, the influenza virus and the single strand plus RNA virus are

preferred, such as hepatitis C virus, dengue virus and Japanese encephalitis virus. Moreover, the

antiviral uses of artificial CpG ODNs and its uses for treating and preventing viral infection are also

provided.

11- Aggregation-free cysteine substitution derivatives of interleukins 28 and 29 retaining

biological activity and their preparation and therapeutic use. Brady, Lowell J.; Klucher, Kevin

M.; Chan, Chung; Dong, Dennis L.; Liu, Hong Y.; Sheppard, Paul O.; Bukowski, Thomas R.

(Zymogenetics, Inc., USA). U.S. Pat. Appl. Publ. (2005), 149 pp. CODEN: USXXCO US

2005037012 A1 20050217 Patent written in English. Application: US 2004-914772 20040809.

Priority: US 2004-559142 20040402; US 2004-551841 20040310; US 2003-493194 20030807.

CAN 142:238656 AN 2005:140553 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Derivs. of interleukins 28 (28A, 28B) and 29 in which a cysteine residue is substituted and which

retain receptor binding and biol. activity are described. The native forms of these interleukins have an

odd no. of cysteines and tend to form intermol. disulfide bonds during purifn. Substitution derivs. do

98

Page 99: dengue

not crosslink and can be purified as monomers. These variants have antiviral activity and may be

useful in the treatment of infection. Manuf. of the variants as inclusion bodies in Escherichia coli

expression hosts using codon-optimized synthetic genes is demonstrated. Biol. activity was tested

using cultured medium from HEK cells showing transient expression of the gene. The conditioned

medium effectively inhibited the cytopathic effect of encephalomyocarditis virus against HeLa cells.

Anal. of gene expression showed that these variants induced the normal interferon response pathway.

12- Sense antiviral oligonucleotide analogs and method for treating ssRNA viral infection.

Iversen, Patrick L. (Avi Biopharma, Inc., USA). PCT Int. Appl. (2005), 81 pp. CODEN:

PIXXD2 WO 2005013905 A2 20050217 Designated States W: AE, AG, AL, AM, AT, AU, AZ, BA,

BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI,

GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, LK, LR, LS, LT, LU,

LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NI, NO, NZ, OM, PG, PH, PL, PT, RO, RU, SC,

SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, ZW.

Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IT, LU, MC, NL, PT, SE,

TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

Application: WO 2004-US25401 20040806. Priority: US 2003-493990 20030807. CAN

142:233275 AN 2005:136499 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The invention provides sense antiviral compds. and methods of their use in inhibition of growth of

virus of the Flaviviridae, Picornoviridae, Caliciviridae, Togaviridae, Coronaviridae families and

hepatitis E virus in the treatment of a viral infection. The sense antiviral compds. are substantially

uncharged morpholino oligonucleotides having a sequence of (12-40) subunits, including at least (12)

subunits having a targeting sequence that is complementary to a region assocd. with stem-loop

secondary structure within the 3'-terminal end (40) bases of the neg.-sense RNA strand of the virus.

13- Preparation of modified fluorinated (2'R)-2'-deoxy-2'-fluoro-2'-C-methyl nucleoside analogs

as antiviral agents. Clark, Jeremy. (Pharmasset, Ltd., Barbados). PCT Int. Appl. (2005), 228

pp. CODEN: PIXXD2 WO 2005003147 A2 20050113 Designated States W: AE, AG, AL, AM, AT,

AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE,

EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, LK, LR,

LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NI, NO, NZ, OM, PG, PH, PL, PT,

RO, RU, SC, SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA,

ZM, ZW. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, IT, LU, MC,

NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent written in English.

99

Page 100: dengue

Application: WO 2004-US12472 20040421. Priority: US 2003-474368 20030530. CAN

142:94074 AN 2005:34765 CAPLUS (Copyright (C) 2010 ACS on SciFinder (R))

Abstract

The disclosed invention provides nucleoside analogs I, wherein B is purine and pyrimidine

nucleobase; X is O, S, CH2, Se, NH, N-alkyl, CHW, C(W)2; W is F, Cl, Br, iodo; R1 is H, phosphate,

H-phosphonate, acyl, Ph, alkyl, carboxyalkylamino, sulfonate ester, peptide, amino acid, sugar reside;

R2 and R2' are independently H, alkyl, alkenyl, alkynyl, vunyl, N3, CN, halogen, NO2, ester, alkoxy,

thioalkyl, sulfoxide, sulfonyl; R6 is alkyl, CN, Me, OMe, OEt, CH2OH, CH2F, N3, CHCN, CH2N3,

CH2NH2, CH2NHMe, CH2NMe2, alkylne; and methods of treating a Flaviviridae infection,

including hepatitis C virus, West Nile Virus, yellow fever virus, and a rhinovirus infection in a host,

including animals, and esp. human, using a (2'R)-2'-deoxy-2'-fluoro-2'-C-Me nucleosides, or a

pharmaceutically acceptable salt or prodrug thereof. Thus, (2'R)-2'-deoxy-2'-fluoro-2'-C-

methylcytidine was prepd. and tested as antiviral agent. The effects the nucleoside analogs tested on

human bone marrow cells are reported. (2'R)-2'-deoxy-2'-fluoro-2'-C-methylcytidine shows activity

against Rhinovirus, West Nile virus, Yellow Fever virus, and Dengue virus. Cytotoxicity and effect of

nucleoside analogs on human bone marrow cells are reported.

14- Motifs of dengue virus envelope protein E of as a novel druggable region for antivirals.

Modis, Yorgo; Harrison, Stephen. (Children's Medical Center Corporation, USA). PCT Int. Appl.

(2005), 123 pp. CODEN: PIXXD2 WO 2005002501 A2 20050113 Designated States W: AE,

AG, AL, AM, AT, AU, AZ, BA, BB, BG, BR, BW, BY, BZ, CA, CH, CN, CO, CR, CU, CZ, DE, DK,

DM, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR,

KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, NA, NI, NO, NZ, OM,

PG, PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SY, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ,

VC, VN, YU, ZA, ZM, ZW. Designated States RW: AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR,

IE, IT, LU, MC, NL, PT, SE, TR, BF, BJ, CF, CG, CI, CM, GA, ML, MR, NE, SN, TD, TG. Patent

written in English. Application: WO 2004-US12433 20040422. Priority: US 2003-464873

20030422; US 2003-505654 20030924. CAN 142:130691 AN 2005:29156 CAPLUS

(Copyright (C) 2010 ACS on SciFinder (R))

Abstract

Motifs of the dengue virus envelope glycoprotein E that may be used as drug targets are identified.

These motifs may be found in other class II viral envelope proteins E and so may form a general target

for virucides. The present invention further relates to methods of using the druggable regions to

screen potential candidate therapeutics for diseases caused by viruses having class II E proteins, e.g.

viral fusion inhibitors. The primary druggable target is the K1 hairpin loop involved in

100

Page 101: dengue

oligomerization of the E protein. Structures of the precursor and sol. forms of the protein were detd.

by X-ray crystallog. Comparison of the structures identified conformational changes and key

structural motifs involved in the oligomerization of the protein in fusion of the virus to the cell

membrane. A hydrophobic ligand-binding pocket was identified in the K1 loop of the protein. This

loop plays an important role in oligomerization and undergoes conformational changes during

formation of the trimer. Peptides derived from the stem region involved in loop formation bound with

fairly high affinity and specificity to the protein.

101