throbber
Therapeutic Advances in Gastroenterology
`
`Review
`
`Hepatitis C virus replication and potential
`targets for direct-acting agents
`
`Jacqueline G. O’Leary and Gary L. Davis
`
`Ther Adv Gastroenterol
`(2010) 3(1) 43—53
`
`DOI: 10.1177/
`1756283X09353353
`! The Author(s), 2010.
`Reprints and permissions:
`http://www.sagepub.co.uk/
`journalsPermissions.nav
`
`Abstract: We finally stand at the brink of novel, oral, direct-acting antivirals for the treatment
`of hepatitis C virus (HCV) infection. Basic science research has lead to a greater understanding
`of the viral life cycle and identified numerous potential targets for therapy. Early compounds
`were plagued by inconsistent in vivo activity and side effects that led to discontinuation of
`investigational efforts. However, several agents have now progressed to phase 2 human
`studies and two protease inhibitors have completed enrolment for their phase 3 clinical trials
`and look promising. Thus, while it appears that protease inhibitors will likely be the next
`available drugs for the treatment of HCV infection, the quest for additional therapeutic agents
`will continue. The future of HCV therapy lies in multidrug cocktails of several agents targeted
`against a variety of targets. In the near future these agents will be added to the current standard
`therapy consisting of pegylated interferon and ribavirin; however, the ultimate and probably
`realistic goal will be to develop multidrug oral regiments to replace the need for interferon.
`
`Keywords: HCV replication, new treatment, polymerase inhibitors, protease inhibitors, therapy
`
`Introduction
`Hepatitis C infects 170 million people worldwide
`and 1.6% of
`the United States population
`[Armstrong et al. 2006; Davis et al. 2003; Alter
`et al. 1999; WHO, 1999]. After acute infection,
`55% to 85% of patients develop chronic disease.
`The natural history of chronic hepatitis C varies
`significantly because of host, viral and environ-
`mental factors. Chronic infection leads to cirrho-
`sis in approximately 20% of patients after 20 years
`of infection [Freeman et al. 2001]. Thereafter,
`other complications including hepatic decompen-
`sation (ascites, encephalopathy, variceal hemor-
`rhage, hepatorenal syndrome, or hepatic synthetic
`dysfunction) and hepatocellular carcinoma ensue
`at a rate of about 3% per year [Sangiovanni et al.
`2006; El-Serag, 2004; Serfaty et al. 1998;
`Fattovich et al. 1997]. Without liver transplanta-
`tion, decompensated cirrhosis leads to death in
`50—72% of patients after 5 years [Fattovich et al.
`2002]. As a result of the high prevalence of hepa-
`titis C virus (HCV) infection and resultant com-
`plications, HCV is the leading indication for liver
`transplantation in the United States and the world
`as a whole [Wasley and Alter, 2000].
`
`Chronic hepatitis C is the only chronic viral
`infection that can be cured with antiviral therapy.
`
`Unlike human immunodeficiency virus and
`hepatitis B, a sustained virologic response
`(SVR), defined as HCV-RNA undetectable by a
`sensitive amplification test 6 months after the
`completion of therapy, is equivalent to a cure in
`>99% of cases [Fried et al. 2002; Manns et al.
`2001]. Patients with compensated cirrhosis who
`achieve an SVR essentially eliminate their subse-
`quent risk of decompensation, may achieve his-
`tologic regression, and decrease their risk of
`hepatocellular carcinoma by two thirds [Bruno
`et al. 2007; Di Bisceglie et al. 2007; Camma
`et al. 2004].
`
`Correspondence to:
`Jacqueline G. O’Leary
`4th Floor Roberts,
`Hepatology-Transplantation,
`Baylor University Medical
`Center,
`3500 Gaston Ave, Dallas,
`TX 75246, USA
`Jacquelo@
`BaylorHealth.edu
`
`Gary L. Davis
`Department of Medicine,
`Baylor University Medical
`Center, Dallas,
`TX, USA
`
`The current standard of care for the treatment of
`HCV infection remains the combination of pegy-
`lated interferon and ribavirin [Fried et al. 2002;
`Manns et al. 2001]. This therapy eradicates HCV
`in 40—50% of genotype 1 non-cirrhotic patients
`and 70—80% of genotype 2 and 3 non-cirrhotic
`patients. The response to treatment is lower in
`obese,
`insulin-resistant, or African-American
`patients and in those with advanced hepatic
`fibrosis or high viral loads. Of greatest concern
`are patients with decompensated cirrhosis and
`immunosuppressed patients, such as liver trans-
`plant recipients, who are rarely able to tolerate
`Gilead 2004
`full doses of therapy.
`I-MAK v. Gilead
`IPR2018-00211
`
`http://tag.sagepub.com
`
`43
`
`

`

`Therapeutic Advances in Gastroenterology 3 (1)
`
`Figure 1. A graphic depiction of the viral life cycle with the potential antiviral targets listed. NS, nonstructural
`proteins; ER, endoplasmic reticulum; IRES, internal ribosome entry side.
`
`Fortunately, we are entering a new era of HCV
`therapy. A greater understanding of the HCV
`replication machinery has led to a large list of
`potential targets for therapy (Figure 1). One
`such target is the nonstructural 3 (NS3) viral
`protease,
`an
`enzyme
`that
`is
`critical
`to
`post-translational processing of the viral polypro-
`tein. Drugs that effectively inhibit this enzyme are
`in the final stages of clinical trials, and it appears
`that the combination of a protease inhibitor with
`pegylated interferon and ribavirin will signifi-
`cantly improve the SVR rate, perhaps even with
`a shorter duration of treatment. In addition,
`these medications will allow us to retreat previous
`relapsers and nonresponders to pegylated inter-
`feron and ribavirin with some success. However,
`while such drugs will be a tremendous addition to
`our therapeutic armamentarium, it is important
`to recognize that they will not eradicate infection
`in all patients and barriers to using interferon or
`ribavirin in many patients will still be a problem.
`
`This article will review the most promising
`potential targets for new direct-acting antiviral
`agents (DAA). Drugs for some of these targets
`are well along in clinical development while
`others are only hypothetical and supported by
`in vitro studies (Table 1). It is important for the
`reader to understand that the high replication
`and nucleic acid substitution rate of HCV will
`
`likely lead to rapid emergence of viral drug resis-
`tance if a single one of these replicative steps is
`targeted. Thus, the future of HCV therapy lies in
`the combination of multiple agents against differ-
`ent targets such as receptor binding, cell entry,
`viral transcription, translation, polyprotein pro-
`cessing, particle assembly and export of virus
`progeny. These DAAs might also be combined
`with non-specific antiviral agents such as those
`that enhance the endogenous immune response
`to the virus, e.g.
`interferons or
`therapeutic
`vaccines, or neutralize extracellular virus, e.g.
`hyperimmune globulins. The goal, of course,
`is to seek combinations that will both increase
`efficacy and improve tolerability.
`
`Early inhibitors: receptor binding and
`cell entry
`Entry of HCV into the hepatocyte involves a
`series of sequential interactions with soluble and
`cell surface host factors, and remains incomple-
`tely understood. Plasma-derived HCV is com-
`plexed with low-density and very-low-density
`lipoproteins (LDL and VLDL), which probably
`facilitates the initial attraction and concentration
`of virus on the cell surface via interaction with
`the low-density lipoprotein receptor (LDL-R)
`[Andre et al. 2005]. The highly glycosylated
`viral
`envelope
`proteins
`E1
`and
`E2
`
`44
`
`http://tag.sagepub.com
`
`

`

`Table 1. Direct-acting agents to treat hepatitis C in phase 2 and 3 clinical trials.
`
`Class
`
`Drug name
`
`Drug Company
`
`JG O’Leary and GL Davis
`
`Entry and RNA-Binding Inhibitors
`Neutralizing Antibodies
`Glycosylation Inhibitors
`
`IRES Inhibitors
`
`Protease Inhibitors
`NS2 Inhibitors
`NS3 Inhibitors
`
`NS4A Inhibitors
`RNA Transcription
`NS4B Inhibitors
`Helicase Inhibitors
`Cyclophilin A Inhibitors
`
`NS5B Polymerase Inhibitors
`
`NS5A Inhibitors
`Other
`TLR-9 Agonists
`NKT Cell Agonist
`Other
`
`Civacir
`Celgosivir
`*UT-231B
`*Heptazyme
`*ISIS-14803
`VGX-410C
`
`none
`Telaprevir
`Boceprevir
`ITMN-191
`TCM435
`MK-7009
`BMS-650032
`BMS-791325
`*ACH-806
`
`none
`none
`Debio-025
`NIM-811
`R7128
`*R1626
`*NM283
`*HCV-796
`VCH-759
`PF-868554
`IDX184
`GS 9190
`BMS790052
`
`*CPG 10101
`KRN7000
`Nitazoxanide
`
`NABI Pharmaceuticals
`Migenix, Inc.
`Unither Pharmaceuticals
`Ribozyme Pharmaceuticals, Inc.
`ISIS Pharmaceuticals
`VGX Pharmaceuticals
`
`Vertex Pharmaceuticals
`Schering-Plough Corporation
`Intermune, Inc.
`Tibotec Pharmaceuticals Limited
`Merck
`Bristol-Myers Squibb
`Bristol-Myers Squibb
`Achillion Pharmaceuticals Inc.
`
`DebioPharm Group
`Novartis
`Pharmasset, Inc. and Roche
`Roche
`Idenix Pharmaceuticals, Inc.
`ViroPharm, Inc.
`Vertex Pharmaceuticals
`Pfizer
`Idenix Pharmaceuticals, Inc.
`Gilead
`Bristol-Myers Squibb
`
`Pfizer
`Kyowa Hakko Kirin Company, Limited
`Romark Laboratories L.C.
`
`Data available at: www.clinicaltrials.gov and www.hcvdrugs.com. NS, nonstructural; IRES, internal ribosome entry site;
`TLR-9, toll-like receptor-9.
`*Future investigation of these compounds has been aborted.
`
`conformationally attach to glycosamioglycans at
`the hepatocyte cell surface and to the c-type lec-
`tins DC-SIGN (dendritic cell-specific intercellu-
`lar adhesion molecule-3-grabbing nonintegrin;
`CD209) and L-SIGN (DC-SIGNr;
`liver and
`lymph node specific; CD209L) on neighboring
`dendritic
`cells
`and
`liver
`sinusoidal
`cells
`[Cormier et al. 2004a]. E2 then sequentially
`attaches to the tetraspanin CD81 and scavenger
`receptor class B type 1 (SR-B1) to form a recep-
`tor complex that utilizes the tight junction clau-
`din
`proteins,
`particularly CLDN1,
`for
`internalization [Zeisel et al. 2007; Cormier et al.
`2004b; Pileri et al. 1998]. Occludin (OCLN), a
`tight junction protein, is also essential for cell
`entry but its exact role remains to be defined
`[Lanford et al. 2009; Ploss et al. 2009; Evans
`et al. 2007]. Interestingly, EWI-2wint, a small
`
`protein that is associated with CD81 in several
`cell
`lines and efficiently blocks HCV entry, is
`absent in hepatocytes and this probably explains
`the selective susceptibility of hepatocytes to HCV
`infection
`[Schuster
`and Baumert,
`2009;
`Rocha-Perugini et al. 2008].
`
`The role of the humoral immune response in con-
`trolling HCV infection is not clear. Monoclonal
`anti-CD81 antibodies have been utilized to effec-
`tively block HCV infection of mice with huma-
`nized livers [Lanford et al. 2009]. However, they
`do not affect HCV infection after it has been
`established. Monoclonal and polyclonal antibo-
`dies with HCV envelope neutralizing capacity
`have been tested in humans at the time of liver
`transplant but have been ineffective in preventing
`reinfection of
`the donor liver; however,
`this
`
`http://tag.sagepub.com
`
`45
`
`

`

`Therapeutic Advances in Gastroenterology 3 (1)
`
`remains a potential perioperative strategy during
`transplantation [Davis et al. 2005]. The currently
`available antibodies’
`lack of efficacy may be
`attributed to the heterogeneity of
`the virus,
`its association with apolipoproteins, or other
`factors.
`
`Another approach to blocking cellular infection is
`to inhibit binding and processing of HCV via the
`cell surface receptor proteins involved in cell
`entry. Glycosylation inhibitors may alter
`the
`structure of cell
`surface glycosaminoglycans
`thereby decreasing or eliminating viral concentra-
`tion at the cell surface [Pawlotsky et al. 2007].
`MX-3256 (Celgosivir; Migenix Inc.) is an oral
`alpha-glucosidase I inhibitor that acts through
`host-directed glycosylation to prevent proper
`folding of the HCV envelope. In preclinical stu-
`dies, celgosivir demonstrated strong synergy with
`pegylated interferon plus ribavirin, but a phase
`IIa monotherapy study did not show any reduc-
`tion in HCV-RNA [Kaita et al. 2007; Yoshida
`et al. 2006]. Unfortunately, a 12-week study of
`another
`glycosylation
`inhibitor, UT-231B
`(Unither Pharmaceuticals),
`in HCV-infected
`patients who previously failed interferon-based
`therapy also failed to reduce virus
`levels.
`Despite these early setbacks, this remains an
`interesting target for future therapies.
`
`In addition, the lectin cyanovirin-N interacts with
`HCV envelope glycoproteins and blocks the asso-
`ciation between E2 and CD81 [Helle et al. 2006].
`Therefore, targeting the cell surface and/or viral
`glycans could be a promising approach to anti-
`viral therapy.
`
`Virus uncoating, HCV-RNA release and
`attachment to the endoplasmic reticulum
`The HCV and receptor complex fuses with the
`hepatocyte
`cell membrane
`and undergoes
`clatharin-mediated endocytosis. Acidification of
`the vesicle leads to fusion of the viral and vesicle
`membranes resulting in uncoating and release of
`the viral RNA into the cytoplasm. The presence
`of viral RNA in the cytosol has pathogenic and
`potential therapeutic implications.
`
`The cell recognizes the presence of single-strand
`RNA (ssRNA) in the cytosol as abnormal and
`initiates a pathogen-associated molecular pattern
`(PAMP)-associated response via toll-like recep-
`tors [Sen, 2001]. This process involves activation
`of
`retinoic
`acid inducible gene-1 (RIG-1)
`and Toll-IL-1 receptor domain containing
`
`that
`adaptor-inducing interferon-beta (TRIF)
`under normal circumstances leads to cellular
`production of
`type 1 interferons and tumor
`necrosis
`factor-related
`apoptosis-inducing
`ligand (TRAIL) mediated apoptosis [Saito and
`Gale, 2008]. However, HCV is capable of down-
`regulating this
`step of
`the innate immune
`response. The HCV NS3/4 protease cleaves and
`inactivates the RIG-1 adaptor protein IFN-beta
`promoter stimulator-1 (IPS-1) and TRIF itself
`thereby blocking downstream activation of the
`interferon regulatory genes [Zhu et al. 2007;
`Foy et al. 2005; Li et al. 2005]. Therefore,
`NS3/4 inhibition (see later), in addition to its
`direct effect on viral polyprotein processing, has
`the potential to restore the RIG-1 and TRIF
`pathways of innate immunity.
`
`The cytosolic viral ssRNA is also a vulnerable
`potential target for therapeutic oligonucleotides
`such as antisense nucleotides (small non-coding
`strands of RNA that hybridize and inactivate
`mRNA) or ribozymes (RNA molecules that cat-
`alyze cleavage of a target RNA). However, these
`agents require an absolutely conserved target in
`an otherwise very heterogeneous virus in order to
`have their effect. The internal ribosomal entry
`site (IRES) at the 5’ end of the viral RNA is
`that highly conserved target. IRES is the landing
`pad that directs the positive strand HCV-RNA to
`the endoplasmic reticulum (ER) for protein
`translation. Thus,
`inhibition of attachment of
`IRES to both cellular and viral proteins by oligo-
`nucleotides could effectively inhibit HCV repli-
`cation. While several class-specific problems with
`oligonucleotides such as drug delivery, instability,
`proinflammatory effects, and other unintended
`‘off-target’ side effects have been partially over-
`come by modifications of the compounds, all
`candidate drugs to date have been plagued by
`safety concerns. Development of Heptazyme
`(Ribozyme Pharmaceuticals, Inc.), a ribozyme
`that cleaved IRES, was stopped secondary to
`toxicity.
`ISIS-14803 (ISIS Pharmaceuticals),
`a 20 base pair antisense oligodeoxynucleotide,
`led to a 1.2 to 1.7 log decline in HCV RNA
`when given as monotherapy three times a week
`for 4 weeks in three out of 28 patients; however,
`asymptomatic liver function test abnormalities
`also
`occurred
`in
`five
`treated
`patients
`[McHutchison et al. 2006]. VGX-410C (VGX
`Pharmaceuticals) was a small molecule that also
`targeted HCV IRES binding. It appeared to be
`safe in phase 2 clinical trials, but was not effec-
`tive. Despite these early setbacks, this approach
`
`46
`
`http://tag.sagepub.com
`
`

`

`JG O’Leary and GL Davis
`
`intriguing
`remains
`exploration.
`
`and warrants
`
`further
`
`Polyprotein translation and protein processing
`Once the viral RNA attaches to the ER, a single
`large polyprotein is translated. This polyprotein
`is then co- and post-translationally processed by
`host and viral proteases into at least 11 viral pro-
`teins. Two host cellular pepsidases are required
`for cleaving HCV structural proteins. These are
`not targets for HCV therapy as they are essential
`for cellular function. NS2 complexes with NS3
`and zinc to form a cystine protease. This complex
`autocatalytically cleaves NS2 from NS3, and is
`then degraded by the proteasome. To date, no
`inhibitors of the NS2 protease have entered clin-
`ical development.
`
`In contrast, the NS3 protease has been the pri-
`mary focus of recent drug development. NS3
`complexes with NS4A and acts as a serine pro-
`tease to cleave the polyprotein at the NS3-NS4A,
`NS4A-NS4B, NS4B-NS5A, and NS5A-NS5B
`sites. The NS3-NS4A complex’s catalytic triad
`lies adjacent to a shallow substrate binding area
`that has made design of potent inhibitors challen-
`ging. It is this active site that is the target for
`drugs
`that are
`currently in clinical
`trials.
`Telaprevir
`(VX-950; Vertex Pharmaceuticals)
`and boceprevir (SCH503034; Schering-Plough
`Corporation) are both in phase 3 clinical trials.
`While these agents are potent inhibitors of HCV
`replication, monotherapy leads to rapid selection
`of drug-resistant strains of the virus, typically
`within days [Kieffer et al. 2007]. Therefore, effec-
`tive treatment, where each drug is dosed three
`times per day, requires combination with other
`agents which means that all current studies
`include pegylated interferon and ribavirin. This
`significantly reduces the likelihood of drug resis-
`tance, but the requirement for frequent dosing
`with these first-generation agents may impede
`compliance and increase the chance of resistance
`outside the setting of clinical trials.
`
`Telaprevir administered during the first 12 weeks
`of a 24 week course of pegylated interferon
`and
`ribavirin
`in
`previously
`untreated
`genotype-1-infected patients resulted in an SVR
`in 61% compared with 41% in those treated with
`pegylated interferon and ribavirin alone for 48
`weeks (standard treatment) [McHutchison et al.
`2008]. A similar trial in Europe achieved an SVR
`in 68% with the 24-week triple-drug regimen
`compared to 48% with standard treatment
`
`[Dusheiko et al. 2008]. A lower dose or elimina-
`tion of ribavirin resulted in a lower chance of
`response. Thus, it appears that, at least for the
`time being, both pegylated interferon and riba-
`virin continue to be essential components of
`treatment. Telaprevir in combination with pegy-
`lated interferon and ribavirin is also effective in
`retreating genotype 1 patients who had relapsed
`or failed to respond to a prior course of pegylated
`interferon
`and
`ribavirin. The
`previously
`described 24 week regimen achieved an SVR in
`69% of prior relapsers and 39% of previous non-
`responders [Manns et al. 2009b]. Extension of
`the total length of treatment to 48 weeks did
`not appear to improve the response to retreat-
`ment. Efficacy and side effects are both increased
`with triple drug therapy. Rash is most common,
`but rarely leads to drug discontinuation. Pruritus,
`nausea, diarrhea and rectal discomfort are also
`more common.
`
`Twenty-eight weeks of therapy with Boceprevir,
`given orally three times a day, in combination
`with peginterferon and ribavirin, led to an SVR
`rate of 55% in previously untreated genotype
`1-infected patients [Kwo et al. 2008]. To address
`concerns about resistance, a second group of
`patients was treated with a 4-week lead-in phase
`of pegylated interferon and ribavirin to reduce
`HCV-RNA levels before the introduction of
`boceprevir; however,
`the SVR rate remained
`unchanged indicating that this is unnecessary.
`Unlike telaprevir, boceprevir
`treated patients
`may benefit from longer therapy. Patients treated
`with 4 weeks of pegylated interferon and ribavirin
`were then treated with either 44 weeks of pegy-
`lated interferon and ribavirin (38% SVR), 24
`weeks of boceprevir, pegylated interferon and
`ribavirin (56% SVR), or 44 weeks of triple ther-
`apy (75% SVR) [Kwo et al. 2009]. Side effects
`with boceprevir include headache, gastrointesti-
`nal complaints and anemia that may limit the
`ability to maintain the ribavirin dose.
`
`ITMN-191 (Intermune, Inc.), another protease
`inhibitor, has been used in combination with
`pegylated interferon and ribavirin for 14 days to
`achieve an undetectable HCV-RNA in 71% of
`treated patients [Kamal and Nasser, 2008].
`ITMN-191 is active against HCV strains that
`are resistant to telaprevir and boceprevir. This
`drug is being studied in combination with pegy-
`lated interferon and ribavirin, as well as in com-
`bination with a polymerase inhibitor (R7128;
`Pharmasset,
`Inc. and Roche) without either
`
`http://tag.sagepub.com
`
`47
`
`

`

`Therapeutic Advances in Gastroenterology 3 (1)
`
`interferon or ribavirin. The latter study is the first
`proof-of-concept study of an interferon-free reg-
`imen in humans. After 14 days of double therapy,
`virus levels had declined by 4.8 to 5.2 logs and
`63%—71% of patients had no detectable virus in
`serum [Gane et al. 2009].
`
`susceptible to resistance, particularly if there is
`not strict adherence to the dosing regimen.
`Therefore, the future of HCV therapy lies with
`improved pharmacodynamics and in combina-
`tions of agents targeting different sites and
`mechanisms of the viral life cycle.
`
`TCM435 (Tibotec Pharmaceuticals Limited and
`Medivir) is a once daily oral NS3 protease inhib-
`itor. A 4.3 to 5.5 log drop (depending on the
`dose) in viral load was achieved after 28 days
`when TCM435 was combined with pegylated
`interferon and ribavirin in genotype 1 HCV-
`infected patients who were previous nonrespon-
`ders or relapsers to interferon based therapy
`[Marcellin et al. 2009]. Three more clinical
`trials utilizing TCM435 with pegylated interferon
`and
`ribavirin
`have
`been
`registered
`at
`ClinicalTrials.gov, two in genotype 1 and one in
`other genotypes.
`
`testing,
`Although in the preclinical phase of
`MK-7009 (Merck) is another NS3 protease
`inhibitor with impressive potency. This drug
`caused a drop of over 5 log10 in HCV RNA in
`5 days in chimpanzees [Lanford et al. 2009].
`When combined with pegylated interferon and
`ribavirin 69 to 82% achieved a rapid virologic
`response compared with 6% who were treated
`with pegylated interferon and ribavirin [Manns
`et al. 2009a]. Like MK-7009, there are many
`other protease inhibitors in various stages of clin-
`ical trials.
`
`Another potential target at the translation step of
`replication is NS4A. NS4A complexes with NS3
`to stabilize the protease functions and anchor the
`protein complex to the endoplasmic reticulum.
`ACH-806 (Achillion Pharmaceuticals Inc.), a
`selective NS4A binder, in vitro caused synergistic
`inhibition of HCV viral replication when used in
`combination with NS3 protease inhibitors such
`as telaprevir [Wyles et al. 2008].
`
`The addition of a protease inhibitor to pegylated
`interferon and ribavirin remains the most prom-
`ising next step in the near future to improve SVR
`rates with HCV therapy. These drugs are potent
`HCV polyprotein processing inhibitors, may
`restore host innate immunity, may improve the
`sensitivity to interferon and are orally bioavail-
`able. These drugs are not without their limita-
`tions, however. They are genotype and probably
`subtype specific to various degrees, have their
`own unique side effects, and will likely remain
`
`HCV-RNA transcription
`Viral transcription occurs in a replicase complex
`built upon a membranous web within the hepa-
`tocyte that is comprised of host and viral ele-
`ments, including the viral polymerase. NS4B is
`essential to construction of the membranous web
`and has important RNA-binding activity facilitat-
`ing attachment of the positive strand viral RNA
`[Egger et al. 2002]. Cyclophilin A recruits NS5B,
`the RNA-dependent RNA polymerase, to the
`viral replication complex. After gathering these
`essential elements at the replication complex,
`the process of strand replication is directed by
`the viral polymerase and generates both the neg-
`ative strand template and positive strand progeny
`to incorporate into new virus particles. The
`newly transcribed positive strand is unwound
`from the template by the viral helicase and this
`single positive sense strand is now available for
`translation, transcription or packaging in new
`virions.
`
`The HCV RNA-dependent RNA polymerase can
`be inhibited by targeting the RNA-binding site or
`one of the other four nonnucleoside allosteric
`sites. Several agents are now in various stages of
`clinical development. R7128 is an active site
`NS5B inhibitor that, in combination with pegy-
`lated interferon and ribavirin, led to an 85% loss
`of detectable virus after just 4 weeks (rapid viro-
`logic response, RVR)
`in naı¨ve genotype 1
`patients. Only 10% of controls receiving pegy-
`lated interferon and ribavirin achieved a RVR
`[Lalezari et al. 2008]. R1626 (Roche), another
`active site NS5B inhibitor, given orally twice a
`day in combination with pegylated interferon
`and ribavirin led to a 74% RVR as compared to
`a 5% RVR in patients treated with pegylated
`interferon and ribavirin [Pockros et al. 2008].
`Unfortunately, R1626 was
`associated with
`dose-limiting
`neutropenia. NM283
`(Idenix
`Pharmaceuticals, Inc.), a third active site inhibi-
`tor of NS5B, was aborted secondary to gastroin-
`testinal side effects.
`
`Several new drugs have also been developed
`against the HCV polymerase’s allosteric sites.
`HCV-796 (ViroPharma,
`Inc.) had entered
`
`48
`
`http://tag.sagepub.com
`
`

`

`JG O’Leary and GL Davis
`
`phase 2 clinical trials in combination with pegy-
`lated interferon and ribavirin; however, further
`study of this drug was discontinued secondary
`to elevated liver
`function tests
`in treated
`patients [Evans et al. 2007]. VCH-759 (Vertex
`Pharmaceuticals), another HCV allosteric inhib-
`itor of the NS5B polymerase, was used in phase 1
`clinical trials as an oral agent dosed three times
`per day [Cooper et al. 2009]. Although drug
`resistance was seen, a 2.5 log10 drop in HCV
`viral
`load was documented after 10 days of
`monotherapy with the highest dose. Pfizer’s com-
`pound, PF-00868554,
`in monotherapy at the
`highest dose resulted in a 1.95 log drop in viral
`load in HCV-infected patients [Hammond et al.
`2008]. Therefore it has progressed to phase 2
`clinical trials.
`
`Polymerase inhibitors represent the second most
`attractive target for HCV therapy after protease
`inhibitors. In addition to the agents described
`above, numerous others are currently in early
`development. Perhaps more than with the pro-
`tease inhibitors, this class of drug has been pla-
`gued with unacceptable side effects that have led
`to discontinuation of investigation of several oth-
`erwise promising compounds. The predominant
`unacceptable side effects have been nausea,
`vomiting and diarrhea, but
`like the protease
`inhibitor side effects, these may be unique to
`each agent rather than class effects. Although
`the nucleoside inhibitors may be genotype spe-
`cific, they may have fewer problems with resis-
`tance than drugs against other
`targets. The
`non-nucleoside
`inhibitors have
`encountered
`rapid emergence of resistance, which may limit
`their usefulness unless they are part of a multi-
`drug cocktail.
`
`There are several potential targets at the step of
`viral transcription other than direct polymerase
`inhibitors. The NS4B, critical for both construc-
`tion of the replicase complex and RNA-binding,
`is a potential target for therapy that has yet to be
`exploited [Lanford et al. 2009]. Cyclophilin inhi-
`bitors, such as Debio-025 (Debiopharm Group)
`and NIM-811 (Novartis), inhibit RNA polymer-
`ase incorporation in the replicase complex.
`Debio-025 is a once-a-day oral agent that has
`already been utilized alone and in combination
`with pegylated interferon and ribavirin in HCV-
`infected patients [Flisiak et al. 2008]. When com-
`bined with pegylated interferon and ribavirin, it
`resulted in a 4.75-log drop in serum HCV RNA
`levels in 29 days, compared with a 2.49-log
`
`decline in HCV RNA levels with pegylated inter-
`feron alone and a 2.2-log decline in HCV RNA
`levels with DEBIO-025 alone. NS5A is a multi-
`function protein that is essential to genome rep-
`lication, particle assembly and the host response
`to the virus [Best et al. 2005]. Its precise role in
`replication is not entirely understood, but
`because it is a multifunctional protein it is an
`extremely attractive target for therapeutic inter-
`vention. Two novel NS5A inhibitors have been
`reported [Lanford et al. 2009]. BMS790052
`(Bristol-Myers Squibb) resulted in a 3.6 log
`decline in HCV RNA following a single dose.
`Like the protease inhibitors, resistance to NS5A
`inhibitors has already been reported. Finally, the
`viral helicase is a potential target. Helicase inhi-
`bitors must be specific for the viral helicase,
`avoiding inhibition of host cellular helicases
`[Dubuisson, 2007; Myong et al. 2007]. No
`HCV helicase inhibitors are currently in clinical
`trials, but preclinical studies of a herpes simplex
`virus helicase inhibitor have shown promise
`[Jankowsky and Fairman, 2007; Kwong et al.
`2005].
`
`Viral assembly and export
`Viral particle formation is initiated by the inter-
`action of the core protein with genomic RNA in
`the endoplasmic reticulum [Mizuno et al. 1995;
`Tanaka et al. 2000]. Viral particle assembly
`requires N-glycosylation of envelope proteins
`for proper envelope folding. This process of enve-
`lope folding and configuration is essential for
`assembly of new virions, virus export, antigeni-
`city
`and receptor binding
`for
`reinfection.
`Therefore, agents that alter envelope glycosyla-
`tion may interfere with numerous steps in the
`viral life cycle (see MX-3256 under ‘Early inhi-
`bitors: receptor binding and cell entry’).
`
`Other
`Interferons have been the cornerstone of hepatitis
`C treatment
`for more than two decades.
`Although interferons stimulate the host innate
`immune response and initiate numerous antiviral
`mechanisms within the cell, the precise effects
`responsible for its efficacy in HCV infection are
`not known. However, some specific host innate
`immune pathways have recently been identified
`and exploited as potential targets for therapeu-
`tics. For
`example,
`two toll-like
`receptor-9
`(TLR-9) agonists are in clinical trials. One such
`compound, CPG 10101 (Pfizer), is a synthetic
`oligodeoxynucleotide that activates TLR-9 lead-
`ing to stimulation of B cells and plasmacytoid
`
`http://tag.sagepub.com
`
`49
`
`

`

`Therapeutic Advances in Gastroenterology 3 (1)
`
`dendritic cells that in turn secrete antiviral cyto-
`kines [McHutchison et al. 2007]. CPG 10101
`when given as monotherapy to HCV-infected
`patients decreased HCV viral
`loads in a dose
`dependent manner, with the highest dose achiev-
`ing a 1.7 log reduction after 4 weeks. One of its
`major effects is to increase cellular interferon.
`Whether this endogenous interferon will prove
`superior to administration of exogenous inter-
`feron is not clear.
`
`KRN7000 (Kyowa Hakko Kirin Company,
`Limited) is another immune activator; specifi-
`cally, a synthetic analog of a-galactosylceramide,
`which stimulates natural killer T cells (NKT)
`in mice and humans. Immune activation of
`NKT cells leads to cytokine production of inter-
`feron-g and tumor necrosis factor-a (TNF-a).
`Although a phase one clinical trial has been com-
`pleted no results have been published (http://
`clinicaltrials.gov/ct2/show/NCT00352235).
`It
`remains unclear if further development of drugs
`that target immune activation will lead to oral
`medications with minimal side effects, or if we
`might simply discover other ways of producing
`cytokines with unacceptable side effects.
`
`NS5A, described above in its role in viral repli-
`cation, also impairs host immune response to
`HCV infection
`in
`numerous ways. The
`N-terminal end of NS5A appears to prevent
`phosphorylation of the Janus Kinases Jak1 and
`Tyk2 and is essential for interferon signaling.
`NS5A also induces the pro-inflammatory cyto-
`kine interleukin-8 (IL-8), which as been asso-
`ciated with an impaired response to interferon
`treatment [Mihm et al. 2004; Polyak et al.
`2001]. In addition, NS5A inhibits apoptosis of
`infected hepatocytes. In other words, NS5A facil-
`itates viral evasion of the host innate immune
`response. In vitro replicon experiments have
`shown that knockout mutations of NS5A result
`in improved interferon sensitivity [Bonte et al.
`2004]. Thus, NS5A inhibitors could enhance
`the sensitivity of infected cells to interferon and
`restore the host’s innate antiviral response.
`
`(Romark Laboratories
`Finally, nitazoxanide
`L.C.) is an antiparasitic agent that was serendi-
`pitously discovered to have antiviral activity
`against hepatitis B and C infection. The pro-
`posed mechanism of action is the increase in
`phosphorylation of the protein kinase PKR that
`phosphorylates eukaryotic initiation factor 2a,
`which leads to the inhibition of HCV-RNA
`
`translation [Darling and Fried, 2009; Rossignol
`et al. 2009; Elazar et al. 2008]. The drug has been
`shown to improve SVR rates in patients infected
`with genotype 4 HCV, the predominant viral gen-
`otype in Egypt where these studies were con-
`ducted. Specifically, 79% of patients achieved
`an SVR when 12 weeks of nitazoxanide was fol-
`lowed by 36 weeks of

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