throbber
Drug Discovery Today: Disease Mechanisms
`
`Vol. 3, No. 4 2006
`
`Editors-in-Chief
`
`Toren Finkel – National Heart, Lung and Blood Institute, National Institutes of Health, USA
`Charles Lowenstein – The John Hopkins School of Medicine, Baltimore, USA
`
`Gastrointestinal disorders
`
`DRUG DISCOVERY
`
`TODAY
`DISEASE
`MECHANISMS
`
`Propagation of hepatitis C virus
`infection: Elucidating targets for
`therapeutic intervention
`David E. Kaplan
`Division of Gastroenterology, University of Pennsylvania, 600 CRB, 415 Curie Blvd., Philadelphia, PA 19104, USA
`
`The hepatitis C virus affects 2–3% of the world’s popu-
`
`lation. The inability to efficiently propagate the virus in
`
`cell-culture greatly impaired the study of the viral life
`
`cycle. Recent advances have facilitated extensive study
`
`of viral entry, translation, proteolysis, and replication
`
`that will foster intelligent drug design for this infection,
`
`a major cause of cirrhosis and hepatocellular carci-
`
`noma worldwide.
`
`Introduction
`The hepatitis C virus (HCV), most commonly via injection
`drug use or blood transfusion, after a generally asymptomatic
`acute infection, persists in 50–75% of exposed subjects. Per-
`sistent infection is associated with chronic hepatitis, which
`progresses to cirrhosis at a rate of approximately 10% per
`decade [1]. Current first-line therapy for chronic hepatitis C
`combines two non-HCV specific medications, interferon-
`alpha (IFNa) injections and oral ribavirin, a nucleoside ana-
`logue. Sustained virological response (SVR), the absence of
`detectable viremia in peripheral blood 6 months after dis-
`continuation of a 24–48 week course of IFNa/ribavirin ther-
`apy, occurs in fewer than half of patients infected with the
`most common genotype of HCV (genotype 1) [2,3]. IFNa/
`ribavirin is associated with significant treatment-related fati-
`gue, depression, hemolytic anemia and other serious side
`effects. Thus, unsatisfactory response rates, poor tolerability
`and frequent adverse events have stirred strong interest in
`
`E-mail address: D.E. Kaplan (dakaplan@mail.med.upenn.edu)
`
`1740-6765/$ ß 2006 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.ddmec.2006.10.004
`
`Section Editor:
`Yu-Xiao Yang – University of Pennsylvania, Philadelphia, USA
`
`specific antiviral therapies, the development of which has
`been recently hastened by advances in in vitro techniques to
`study HCV viral replication.
`
`The virus
`The hepatitis C virus (HCV) is a highly mutable, hepatotropic,
`enveloped 9.5 kB single-stranded RNA virus of the Flavivir-
`idae family that can infect humans and chimpanzees. HCV has
`high sequence heterogeneity, with at least 6 distinct genotypes
`and at least 50 subtypes distributed worldwide [4]. Within
`individual subjects, HCV exists as a swarm of closely related
`variants (quasispecies); this variation results from a highly
`error prone RNA polymerase that lacks proofreading capacity.
`While most mutations are deleterious, a small percentage of
`random mutations confer upon nascent viral strains a selective
`replication advantage, which fosters continual viral adapta-
`tion to the unique and dynamic environment provided by the
`host, a process that plays a major role in drug resistance under
`conditions of sub-maximal viral suppression.
`Hepatitis C, like all viruses, must be capable of entering
`target cells, transcribing and/or translating the genetic mate-
`rial to produce structural and nonstructural components for
`new virion formation, replicating the genetic material to
`new include in nascent virions, packaging the genetic
`material into the virion structure, and releasing daughter
`viruses (Fig. 1). Characterization of the critical steps of
`HCV replication was initially hampered by the inability to
`(cid:42)(cid:76)(cid:79)(cid:72)(cid:68)(cid:71)(cid:3)(cid:21)(cid:19)(cid:19)(cid:22)(cid:3)
`(cid:44)(cid:16)(cid:48)(cid:36)(cid:46)(cid:3)(cid:89)(cid:17)(cid:3)(cid:42)(cid:76)(cid:79)(cid:72)(cid:68)(cid:71)(cid:3)
`(cid:44)(cid:51)(cid:53)(cid:21)(cid:19)(cid:20)(cid:27)(cid:16)(cid:19)(cid:19)(cid:22)(cid:28)(cid:19)
`
`471
`
`

`

`Drug Discovery Today: Disease Mechanisms | Gastrointestinal disorders
`
`Vol. 3, No. 4 2006
`
`Figure 1. Hepatitis C viral lifecycle. (i) Cell entry is mediated by E1 and E2 glycoprotein interacting with cellular receptors such as CD81 and HDL
`scavenger receptor type B1, which triggers receptor-mediated endocytosis. (ii) The virus uncoats, leaving positive-strand RNA in cytoplasm. (iii) RNA
`interacts with ribosome on endoplasmic reticulum (ER) via internal ribosome entry site (IRES) leading to translation of large polyprotein. (iv) Host and viral
`enzymes cleave the polyprotein into individual proteins. (v) Viral proteins lead to organization of membranous web (replication complex). (vi) NS5B and
`NS3 helicase along with host factors catalyze transcription of negative-strand RNA template, then replication of positive-strand copies with temporarily
`double-strand RNA intermediate.
`
`grow wild-type HCV in culture and the lack of widely avail-
`able small animal models to study infection in vivo. Most
`investigation of HCV protein function and replication has
`resulted from use of recombinant viral constructs that express
`HCV proteins under the control of promoters, internal ribo-
`some entry sites (IRES), or nonstructural proteins borrowed
`from unrelated viruses. These constructs (replicons, retroviral
`pseudoparticles, chimeric viruses) recapitulated some but not
`all steps of the viral life cycle in vitro, but often required
`specific adaptive mutations and/or highly adapted host cell
`lines (reviewed in [5,6]). In 2005, JFH1, an infectious clone of
`HCV capable of replication in human hepatoma cell lines and
`chimpanzee hepatocytes [7], was characterized. Combined,
`the various in vitro model systems have markedly advanced
`the understanding of viral propagation and spurred pre-
`
`clinical development of a variety of specific inhibitors target-
`ing critical steps in viral replication (Table 1).
`
`Characterization and inhibition of HCV entry into
`permissive host cells
`Unlike HIV infection in which the cellular receptor for the viral
`envelope proteins is well characterized and has spawned devel-
`opment of fusion inhibitors, the mechanism by which HCV
`enters target cells remains incompletely characterized. The
`circulating virion includes the a nucleocapsid consisting of
`the HCV Core protein and positive-strand ssRNA surrounded
`by an envelope, which consists of a lipid bilayer derived from
`the previous host cell’s membrane and HCV envelope glyco-
`proteins E1 and E2. E1 and E2 form heterogeneous mixtures of
`covalently- and noncovalently-associated heterodimers that
`
`472
`
`www.drugdiscoverytoday.com
`
`

`

`Vol. 3, No. 4 2006
`
`Drug Discovery Today: Disease Mechanisms | Gastrointestinal disorders
`
`Table 1. Viral and host cellular protein targets for specific inhibitor therapya
`
`Therapy against
`target
`
`Stage of
`development
`
`Advantages and/or
`disadvantages
`
`NS3/4 protease
`
`Peptidomimetic
`
`Phase II
`
`Potent as monotherapy– 3–4 log10 drop;
`viral escape; strain-specificity
`
`NS5B polymerase
`
`Nucleotide analogue
`Non-nucleotide inhibitors
`(benzimidazole, benzothiadiazine,
`thiophene caroboxylic acids)
`
`Phase III
`
`Low potency as monotherapy?
`Adverse effects?
`Viral escape
`
`NS3 helicase
`
`IRESb
`
`Modified benzimidazoles,
`ribavirin esters,
`tropolone analogues
`
`Antisense
`shRNA
`siRNA
`Ribozyme
`Aptamers
`Peptide-nucleic acids
`Other small molecule
`
`Phase I?
`
`Unknown effects in vivo
`
`Phase II
`
`Delivery mechanism
`Toxicity?
`Potentially more resistant to
`viral escape due to conservation
`of IRES
`Cross-genotype activity
`
`Cell entry
`(CD81, SR-B1c,
`d
`DC-SIGN
`)
`
`Particle formation
`(lipids, glycoproteins)
`
`Blocking antibody
`(endogenous, exogenous)
`Small molecules
`
`Alpha-glucosidase inhibitors
`Prenylation inhibitors
`Sphingolipidation blockers
`
`Phase I
`
`Strain specificity
`
`Viral escape
`
`Pre-clinical
`
`Unknown
`
`a Published safety and efficacy data are very limited—advantages/disadvantages are highly speculative.
`b Internal ribosome entry site.
`c HDL scavenger receptor type B1.
`d Dendritic cell-specific ICAM3 grabbing nonintegrin.
`
`Who is working on
`the target (group and
`institute or company)
`
`Refs
`
`Vertex
`
`Schering-Plough
`Gilead
`Intermune
`Others
`
`Idenix
`Roche
`XTL
`Pharmasset
`Viropharma
`Others
`
`Vertex
`
`Viral Genomix
`AVI Biopharma
`Others
`
`[25,26]
`
`[30,31]
`
`[33,34]
`
`[18,19,23]
`
`Innogenetics
`XTL
`Others
`
`Migenix
`
`[13]
`
`[36,38–41]
`
`mediate viral entry. In cell culture systems, mainly utilizing
`viral pseudoparticles and more recently the JFH1 clone, viral
`entry occurs via low-pH-dependent, clathrin-mediated recep-
`tor-mediated endocytosis [8]. The E2 glycoprotein mediates
`binding to cellular receptors such as CD81, a tetraspannin
`receptor expressed on hepatocytes among other cell types,
`and the HDL receptor scavenger receptor class B type 1 (SR-
`B1), blockade of which inhibits E2-mediated viral entry in vitro
`[9,10]. Other putative co-receptors including the LDL-receptor,
`C-type lectins, L-SIGN and DC-SIGN [11], may be involved.
`Recent data identifies a conserved GWLAGLFY motif at posi-
`tion 436 of E2 as the critical residues required for CD81-
`dependent viral entry [12]. The accessibility of virus-cell sur-
`face interactions allows consideration for antibody-based, pep-
`tidomimetic or small molecule approaches to disrupt early
`stages of cellular infection.
`Some preliminary success has been reported with envelope
`protein-including vaccines [13] (INN-0101, Innogenetics,
`Gent, Belgium, http://www.innogenetics.com/) and with
`human monoclonal antibodies against E2 (XTL-6865, XTL,
`
`Rehovot, Israel, http://www.xtlbio.com/) to augment clear-
`ance of circulating viral particles and/or inhibit cellular re-
`infection. A polyclonal immune globulin is also in develop-
`ment (Civacir, Nabi Pharmaceuticals, Boca Raton, FL USA,
`http://www.nabi.com/). At this time, data regarding specific
`small molecule inhibitors of E2-CD81 or other envelope-cell
`receptor binding have yet to be published.
`
`Genome translation
`Once HCV has entered a target cell, translation of the viral
`positive strand RNA proceeds using host cell machinery. The
`9.6 kB RNA genome encodes a single open reading frame
`0
`0
`and 3
`non-coding regions. Three hundred
`(ORF) flanked by 5
`0
`untranslated region (and begin-
`thirty nucleotides of the 5
`ning of the core protein encoding nucleotides) of the virus
`contain the internal ribosome entry site (IRES), a highly
`structured domain critical for initiation of HCV polyprotein
`translation. The HCV IRES contains
`three stem-loops
`(domains II–IV) and a pseudoknot [14]. Translation mediated
`0
`by the HCV IRES eliminates the needs for a 5
`cap structure on
`
`www.drugdiscoverytoday.com
`
`473
`
`

`

`Drug Discovery Today: Disease Mechanisms | Gastrointestinal disorders
`
`Vol. 3, No. 4 2006
`
`RNA, ATP-dependent scanning, and multiple canonical
`initiation factors (reviewed in [15]). The human La autoanti-
`gen appears required for the interaction of the IRES and the
`40S ribosome subunit [16]. Due to constraints on stem-loop
`base-pairing, only a minority of nucleotides can be substi-
`tuted without impairment IRES functionality and thus the
`region is highly conserved among HCV subtypes [17]. These
`constraints similarly make the IRES an attractive target for
`therapeutics.
`Initial approaches to inhibit IRES function have focused on
`0
`antisense oligonucleotides to block the interaction of the 5
`IRES with the 40S ribosome [18] and two agents are currently
`in phase I/II trials (VGX-410C, Viral Genomix Inc. Blue Bell,
`PA, USA, http://www.viralgenomix.com/; AVI-4065, AVI Bio-
`pharma, Portland, OR, USA, http://www.avibio.com/). ISIS
`14803 (ISIS Pharmaceuticals, Carlsbad, CA, USA, http://
`www.isispharm.com/) showed some promise but questions
`regarding toxicity in early development [19] and was sus-
`pended. Other RNA-based IRES inhibitors such as RNAi,
`shRNA, ribozymes, aptamers, and peptide nucleic acids that
`disrupt IRES function have been shown to inhibit in vitro
`replication of HCV RNA [20,21]. A caveat to this approach is
`that there is some evidence that HCV core protein interferes
`with siRNA activity [22]. A novel competitive La peptide
`variant [23] has been shown in vitro to impair HCV replication
`via IRES inhibition and may be a candidate for further devel-
`opment.
`
`Protein processing
`After IRES engagement with ribosomes, viral RNA is trans-
`lated as a single long polyprotein which requires proteolytic
`cleavage (Fig. 2). Host cell signal peptidases cleave at the
`Core/E1, E1/E2, E2/p7 and p7/NS2 junctions. The Core pro-
`tein forms the viral nucleocapsid which localizes replicated
`
`RNA via a highly conserved RNA binding domain, with the
`budding envelope studded with E1 and E2 heterodimers. The
`p7 protein encodes an ion channel critical for viral infectiv-
`ity. The NS2/NS3 cleavage is catalyzed by NS2, a transmem-
`brane zinc-dependent protease, a step critical to efficient
`function of the NS3 protein; NS2 may also regulate certain
`cellular factors involved with apoptosis and fibrosis. The
`remaining cleavages, those at NS3/NS4, NS4A/NS4B, NS4B/
`NS5A, and NS5A/NS5B, are catalyzed by the NS3 using NS4A
`as a cofactor, termed the NS3/4A serine protease. Addition-
`ally, NS3 has a separate NTP-dependent RNA helicase
`domain. NS4A is a cofactor for the NS3 protease and recent
`data indicate that the NS3/4A protease blocks signaling path-
`ways that trigger NF-kB nuclear translocation and interferon-
`response factor pathways by cleaving an adaptor protein
`necessary for RIG-I signaling, called Cardif (also known as
`MAVS, IPS-1 and VISA) [24]. The role of NS4B is unclear, but it
`may along with NS4A regulate viral translation and replica-
`tion. The NS5A protein contains a PKR binding region and
`may modulate sensitivity to interferon-alpha and NS5B is the
`viral RNA-dependent RNA polymerase.
`As described,
`the NS3 protein has three functional
`domains: an autoprotease that cleaves the NS2/3 junction
`using NS2 as a cofactor, an RNA helicase domain and the NS3/
`4A protease. The NS3/4A serine protease in addition to its
`proteolytic function also appears to regulate the NS3 helicase
`domain. Due to the critical role that the NS3/4 protease plays
`in various aspects of viral replication, development of a
`specific inhibitor has been a therapeutic priority. BILN
`2061 (Boehringer Ingelheim GmbH, Ingelheim, Germany,
`http://www.boehringer-ingelheim.com/), the first of these
`inhibitors to enter clinical trials achieved 2–3 log10 reduc-
`tions in HCV RNA titers [25] in genotype 1 infections. Both in
`vitro and in vivo BILN 2061 was significantly less effective
`
`0
`UTR (untranslated region) contains the
`Figure 2. Hepatitis C viral translation and proteolysis. Translation and proteolysis of the HCV polyprotein. 5
`internal ribosome entry site, which facilitates interaction of positive-strand RNA with the 40S ribosome resulting in translation of a single polyprotein of
`3011 amino acids. Numerals indicate amino acid positions for subsequent cleavages. Blue triangles indicate cleavages catalyzed by host cell signal peptide
`peptidases. The green triangle shows cleavage site of NS2/3 autoprotease. Red triangles indicate cleavage sites for the NS3/4A protease.
`
`474
`
`www.drugdiscoverytoday.com
`
`

`

`Vol. 3, No. 4 2006
`
`Drug Discovery Today: Disease Mechanisms | Gastrointestinal disorders
`
`against genotype non-1 NS3/4A proteases. Development of
`BILN 2061 was suspended due to cardiotoxicity in non-
`human primates given the drug. VX-950 (Vertex Pharmaceu-
`ticals, Cambridge MA USA, [26], http://www.vpharm.com/)
`and SCH 503034 (Schering-Plough, Kenilworth NJ USA,
`http://www.schering-plough.com/) are currently in phase II
`and at least two more candidates (GS 3192/ACH806, Gilead,
`Foster City CA USA, http://www.gilead.com/; ITMN 191,
`Intermune, Brisbane CA, USA, http://www.intermune.com/)
`are in phase I testing with a host of other candidate drugs
`nearing clinical testing. Published safety, tolerability, and
`efficacy data for this class of drugs are limited, but VX-950
`administered orally three times daily led to median 3 log10
`reductions in HCV RNA after 3 days and 4.4 log10 reductions
`after 14 days [26] in a phase II trial.
`
`The NS3 helicase enzymatically unwinds duplex RNA
`structures by disrupting the hydrogen bonds that keep the
`two strands together, an activity that requires hydrolysis of
`nucleoside triphosphate (NTP). Potential specific inhibitors
`of NTPase/helicases theoretically could function by inhibit-
`ing NTP binding or NTPase activity, uncoupling NTP hydro-
`lysis from the unwinding reaction, competing with dsRNA
`binding and/or by blocking translocation of the NTPase/heli-
`case along the dsRNA and thus blocking propagation of
`unwinding [32]. Peptide libraries have identified aminophe-
`nylbenzimidazole moieties [32],
`imidazo[4,5-d]pyridazine
`nucleosides, halogenated benzimidazoles, halogenated ben-
`0
`-O-(4-fluorosulphonylbenzoyl)-esters of riba-
`zotriazoles, 5
`virin [33], and bromo- and morpholinomethyl-analogues
`of tropolone [34] as potential inhibitors of the NS3 helicase.
`
`Genomic replication
`The primary goal of the viral proteins is to coordinate the
`replication of viral RNA and the packaging new RNA strands
`into nascent structural units that can be extruded. As a
`positive-strand RNA virus, RNA replication depends on the
`creation of a negative-strand RNA template, from which new
`positive-strands can be duplicated. RNA replication occurs in
`the cytoplasm facilitated by the formation of a membrane-
`associated complex involving all of the nonstructural viral
`proteins as well as host factors [27]. Two viral enzymes
`play the dominant roles in this process: the NS5B RNA, the
`RNA-dependent RNA polymerase and the NS3 helicase.
`The NS5B polymerase is a right hand structured polymerase
`with thumb, palm and finger domains and a completely
`encircled active site [28], for which both nucleotide and
`non-nucleoside inhibitors have been identified. Nucleotide
`analogs compete with nucleotide binding at the polymerase
`active site and can cause chain termination upon incorpora-
`tion into the RNA molecules [29]. Non-nucleoside inhibitors of
`NS5B RNA synthesis activity mainly consist of a heterogeneous
`group of benzimidazole, benzothiadiazine, and thiophene
`carboxylic-acid-based compounds that bind to three non-over-
`lapping binding sites on NS5B [30] and work primarily by
`allosterically inhibiting formation of replicase complexes.
`There are at least five RNA polymerase inhibitors in phase I–
`III human clinical testing at present: NM283 (Idenix, Cam-
`bridge MA, USA, http://www.idenix.com/), R1626 (Roche,
`Nutley NJ USA, http://www.roche.com/), XTL-2125 (XTL,
`Rehovot, Israel), PSI-6130 (Pharmasset Inc., Princeton, NJ
`USA http://www.pharmasset.com/) and HCV-796 (Viro-
`pharma, Exton PA USA, http://www.viropharma.com/). Cur-
`rently, safety and tolerability data are limited, but preliminary
`0
`results from NM283, a prodrug of 2
`-C-methylcytidine, indi-
`cate a fairly modest reductions in HCV RNA titers (<1 log10
`decrease)
`for monotherapy but virologic response rates
`similar to interferon/ribavirin when used in combination
`with interferon [31].
`
`Inhibitors of viral particle formation
`In order for new virions to be completed, nascent positive
`strand HCV RNA must be packaged into structural units. The
`replication of HCV RNA occurs in membrane-associated
`complexes, and the structural units are assembled closely
`associated with membranes. This membrane association
`depends on glycosylation of proteins and lipophilic protein
`modifications, processes that could be considered for thera-
`peutic interference.
`The importance of lipids and HCV was first suggested by
`the interaction of HCV with the LDL receptor and/or HDL
`scavenger receptor B1. Evidence now suggests that HMG-coA
`(fluvastatin > atorvastatin,
`reductase
`inhibitors
`lovasta-
`tin > simvastatin) do indeed have anti-HCV activity [35],
`but surprisingly that this inhibition more likely occurs at
`the level of viral replication, not at the level of cell entry, and
`that these effects are reversed by the addition of cholesterol
`biosynthesis metabolites [35–37]. Studies in HCV replicon
`systems further demonstrate that HCV replication is intri-
`cately controlled by fatty acid and cholesterol metabolism;
`HCV RNA replication is inhibited in vitro with polyunsatu-
`rated fatty acids and acetyl-coA carboxylase inhibitors [37]. In
`culture, the combination of interferon-alpha and fluvastatin
`exhibited strong synergistic inhibitory effects on HCV RNA
`replication suggesting that fluvastatin in particular, but
`potentially other statins, could be potentially useful as an
`adjunct to interferon-alpha [35].
`An offshoot of cholesterol metabolism, the prenylation
`pathway, also appears to be important in viral particle assem-
`bly for the unrelated hepatitis D virus [38,39] and the findings
`from the HCV replicon system also suggest dependence of
`HCV particle production on a prenylated substrate [36].
`Geranylgeranyl diphosphate is derived from farnesyl dipho-
`sphate itself a product of the cholesterol biosynthesis path-
`way (Fig. 3). Farnesylation and geranylgeranylation (together
`termed prenylation) are catalyzed by specific enzymes (far-
`nesyl transferase, geranylgeranyl transferase I and II) for
`
`www.drugdiscoverytoday.com
`
`475
`
`

`

`Drug Discovery Today: Disease Mechanisms | Gastrointestinal disorders
`
`Vol. 3, No. 4 2006
`
`Figure 3. Cholesterol biosynthesis pathways and potential enzymatic targets affecting HCV RNA replication. Farnesylation geranylgeranylation signal
`sequences consist of CXXK and CXXL motifs, respectively, resulting in lipidation of cysteine residues and cleavage of XXK and XXL residues. Lipidation
`fosters membrane localization and possibly interaction with the multiple-drug resistance (MDR) transporter.
`
`which specific small molecule inhibitors exist. Prenylation
`fosters membrane association of the modified target protein
`and might also facilitate extrusion from the cell via the
`multiple drug resistance transporter. Although the specific
`proteins of HCV that are dependent on prenylation for their
`function have yet to be determined, theoretically inhibition of
`prenylation might be another avenue for drug development in
`HCV. Recently, a sphingolipid modification of NS5B necessary
`for linking NS5B to lipid rafts, a step thought to be critical for
`the association of NS5B with other membrane bound viral
`proteins, was found to be sensitive to inhibition in vitro by a
`secondary fungal metabolite [40]. Human cellular alpha-glu-
`cosidase blockade has been shown to inhibit glycosylation of
`various HCV proteins and may inhibit viral particle assembly
`[41]; Celgosivir (MBI-3253, Migenix, Vancouver BC Canada,
`http://www.migenix.com/), an oral prodrug of castanosper-
`mine, a natural product derived from the Australian Black
`Bean chestnut tree, targets this host cell enzyme and may
`have efficacy. Thus, the interaction of viral proteins and
`subcellular membranes represents a relatively novel area
`for development.
`
`Conclusion
`Specific antiviral therapies for hepatitis C will fill a critical
`unmet demand for patients with chronic HCV that have
`either not responded to, are unable to receive, or are unable
`to tolerate interferon-based therapy. Other types of therapy
`in active development, but beyond the scope of this review,
`include novel or improved immunomodulators such as TLR7
`
`476
`
`www.drugdiscoverytoday.com
`
`agonists, thymosin, modified interferons, and therapeutic
`vaccines. Although invaluable for initial preclinical develop-
`ment, the remarkable advances in HCV in vitro culture sys-
`tems cannot foreshadow the effects of these agents in vivo due
`to various confounders such as unexpected toxicities, diffi-
`culties in drug delivery, alternative in vivo metabolism, and
`perhaps most importantly sensitivity to viral resistance. The
`rapid mutation rate of HCV will clearly necessitate combina-
`tions of complementary direct antivirals with or without
`concomitant interferon-alpha (or other immunomodula-
`tors). Due to limited availability of safety and efficacy data
`for most of the agents in development, predictions as to when
`specific antivirals will enter the clinical armamentarium
`against HCV remain difficult; a five-year horizon would be
`optimistic. However, the wide variety of potential targets
`elucidated by in vitro studies raises the likelihood that one
`or several of the strategies under investigation will yield
`significant clinical benefits for the huge populations of
`patients worldwide infected with the hepatitis C virus.
`
`References
`1 Seeff, L.B. (2002) Natural history of chronic hepatitis C. Hepatology 36, S35–
`S46
`2 Manns, M.P. et al. (2001) Peginterferon alfa-2b plus ribavirin compared
`with interferon alfa-2b plus ribavirin for initial treatment of chronic
`hepatitis C: a randomised trial. Lancet 358, 958–965
`3 Fried, M.W. et al. (2002) Peginterferon alfa-2a plus ribavirin for chronic
`hepatitis C virus infection. N. Engl. J. Med. 347, 975–982
`4 Simmonds, P. et al. (1993) Classification of hepatitis C virus into six major
`genotypes and a series of subtypes by phylogenetic analysis of the NS-5
`region. J. Gen. Virol. 74, 2391–2399
`
`

`

`Vol. 3, No. 4 2006
`
`Drug Discovery Today: Disease Mechanisms | Gastrointestinal disorders
`
`5 Guha, C. et al. (2005) Cell culture models and animal models of viral
`hepatitis. Part II: hepatitis C. Lab. Anim. (NY) 34, 39–47
`6 Brass, V. et al. (2006) Molecular Virology of Hepatitis C Virus (HCV): 2006
`Update. Int. J. Med. Sci. 3, 29–34
`7 Wakita, T. et al. (2005) Production of infectious hepatitis C virus in tissue
`culture from a cloned viral genome. Nat. Med. 11, 791–796
`8 Blanchard, E. et al. (2006) Hepatitis C virus entry depends on clathrin-
`mediated endocytosis. J. Virol. 80, 6964–6972
`9 McKeating, J.A. et al. (2004) Diverse hepatitis C virus glycoproteins mediate
`viral infection in a CD81-dependent manner. J. Virol. 78, 8496–8505
`10 Barth, H. et al. (2005) Scavenger receptor class B type I and hepatitis C virus
`infection of primary tupaia hepatocytes. J. Virol. 79, 5774–5785
`11 Lozach, P.Y. et al. (2004) C-type lectins L-SIGN and DC-SIGN capture and
`transmit infectious hepatitis C virus pseudotype particles. J. Biol. Chem.
`279, 32035–32045
`12 Drummer, H. et al. (2006) A conserved Gly436-Trp-Leu-Ala-Gly-Leu-Phe-
`Tyr motif in hepatitis C virus glycoprotein E2 is a determinant of CD81
`binding and viral entry. J. Virol. 80, 7844–7853
`13 Nevens, F. et al. (2003) A pilot study of therapeutic vaccination with
`envelope protein E1 in 35 patients with chronic hepatitis C. Hepatology 38,
`1289–1296
`14 Honda, M. et al. (1996) Structural requirements for initiation of translation
`by internal ribosome entry within genome-length hepatitis C virus RNA.
`Virology 222, 31–42
`15 Finotto, S. et al. (2004) T cell directives for transcriptional regulation in
`asthma. Springer Semin. Immunopathol. 25, 281–294
`16 Pudi, R. et al. (2003) Hepatitis C virus internal ribosome entry
`site-mediated translation is stimulated by specific interaction of
`independent regions of human La autoantigen. J. Biol. Chem. 278,
`12231–12240
`0
`17 Simmonds, P. et al. (1993) Sequence variability in the 5
`non-coding region
`of hepatitis C virus: identification of a new virus type and restrictions on
`sequence diversity. J. Gen. Virol. 74 (Pt 4), 661–668
`18 Soler, M. et al. (2004) Virological effects of ISIS 14803, an antisense
`oligonucleotide inhibitor of hepatitis C virus (HCV) internal
`ribosome entry site (IRES), on HCV IRES in chronic hepatitis C patients
`and examination of the potential role of primary and secondary
`HCV resistance in the outcome of treatment. Antivir. Ther. 9,
`953–968
`19 McHutchison, J.G. et al. (2006) A phase I trial of an antisense inhibitor of
`hepatitis C virus (ISIS 14803), administered to chronic hepatitis C patients.
`J. Hepatol. 44, 88–96
`20 Romero-Lopez, C. et al. (2005) Interfering with hepatitis C virus IRES
`activity using RNA molecules identified by a novel in vitro selection
`method. Biol. Chem. 386, 183–190
`21 Liang, X.S. et al. (2004) Inhibitor RNA blocks the protein translation
`mediated by hepatitis C virus internal ribosome entry site in vivo. World J.
`Gastroenterol. 10, 664–667
`22 Wang, Y. et al. (2006) Hepatitis C virus core protein is a potent inhibitor
`of RNA silencing-based antiviral response. Gastroenterology 130,
`883–892
`23 Pudi, R. et al. (2005) A peptide derived from RNA recognition motif 2 of
`human la protein binds to hepatitis C virus internal ribosome entry site,
`
`prevents ribosomal assembly, and inhibits internal initiation of
`translation. J. Virol. 79, 9842–9853
`24 Sumpter, R., Jr et al. (2005) Regulating intracellular antiviral defense and
`permissiveness to hepatitis C virus RNA replication through a cellular RNA
`helicase, RIG-I. J. Virol. 79, 2689–2699
`25 Hinrichsen, H. et al. (2004) Short-term antiviral efficacy of BILN 2061, a
`hepatitis C virus serine protease inhibitor, in hepatitis C genotype 1
`patients. Gastroenterology 127, 1347–1355
`26 Gelderblom, H.C. (2005) . In Decline in Serum Neopterin Concentration
`Correlates with HCV RNA Decline During Administration of VX-950, a
`Hepatitis C Virus Protease Inhibitor (Zeuzem, S. et al. eds), p. 859A, Vertex
`Pharmaceuticals
`27 Moradpour, D. et al. (2003) Membrane association of hepatitis C virus
`nonstructural proteins and identification of the membrane alteration that
`harbors the viral replication complex. Antiviral Res. 60, 103–109
`28 Lesburg, C.A. et al. (1999) Crystal structure of the RNA-dependent RNA
`polymerase from hepatitis C virus reveals a fully encircled active site. Nat.
`Struct. Biol. 6, 937–943
`29 Carroll, S.S. et al. (2006) Nucleoside analog inhibitors of hepatitis C virus
`replication. Infect. Disord. Drug Targets 6, 17–29
`30 Ma, H. et al. (2005) Inhibition of native hepatitis C virus replicase by
`nucleotide and non-nucleoside inhibitors. Virology 332, 8–15
`31 Afdhal, N. et al. (2005) Enhanced antiviral efficacy for
`valopicitabine(NM283) plus peg-interferon in hepatitis C patients with
`HCV genotype 1 infection: Results of a phase IIa multicenter trial.
`European Association for the Study of the Liver, Annual Meeting 2005, Paris,
`France
`32 Borowski, P. et al. (2002) Nucleotide triphosphatase/helicase of
`hepatitis C virus as a target for antiviral therapy. Antiviral Res. 55,
`397–412
`33 Phoon, C.W. et al. (2001) Biological evaluation of hepatitis C virus helicase
`inhibitors. Bioorg. Med. Chem. Lett. 11, 1647–1650
`34 Boguszewska-Chachulska, A.M. et al. (2006) Searching for a new anti-HCV
`therapy: synthesis and properties of tropolone derivatives. Biochem.
`Biophys. Res. Commun. 341, 641–647
`35 Ikeda, M. et al. (2006) Different anti-HCV profiles of statins and
`their potential for combination therapy with interferon. Hepatology 44,
`117–125
`36 Ye, J. et al. (2003) Disruption of hepatitis C virus RNA replication through
`inhibition of host protein geranylgeranylation. Proc. Natl. Acad. Sci. U S A
`100, 15865–15870
`37 Kapadia, S.B. et al. (2005) Hepatitis C virus RNA replication is regulated by
`host geranylgeranylation and fatty acids. Proc. Natl. Acad. Sci. U S A 102,
`2561–2566
`38 Einav, S. et al. (2003) Prenylation inhibitors: a novel class of antiviral
`agents. J. Antimicrob. Chemother. 52, 883–886
`39 Glenn, J.S. et al. (1998) Use of a prenylation inhibitor as a novel antiviral
`agent. J. Virol. 72, 9303–9306
`40 Sakamoto, H. et al. (2005) Host sphingolipid biosynthesis as a target for
`hepatitis C virus therapy. Nat. Chem. Biol. 1, 333–337
`41 Chapel, C. et al. (2006) Antiviral effect of alpha-glucosidase inhibitors on
`viral morphogenesis and binding properties of hepatitis C virus-like
`particles. J. Gen. Virol. 87, 861–871
`
`www.drugdiscoverytoday.com
`
`477
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket