throbber
Review
`
`Antiviral Chemistry & Chemotherapy 16:69–90
`
`Host cell targets in HCV therapy: novel strategy or
`proven practice?
`Bert M Klebl1*, Alexander Kurtenbach1, Kostas Salassidis1, Henrik Daub1 and Thomas Herget2
`
`1Axxima Pharmaceuticals AG, Munich, Germany
`2New Business Chemicals Bio, Merck KGaA Germany, Darmstadt, Germany
`
`*Corresponding author: Tel: +49 89 55065 460; Fax: +49 89 55065 461; E-mail: bert.klebl@axxima.com
`
`The development of novel antiviral drugs against
`hepatitis C is a challenging and competitive area of
`research. Progress of this research has been
`hampered due to the quasispecies nature of the
`hepatitis C virus, the absence of cellular infection
`models and the lack of easily accessible and highly
`representative animal models. The current combi-
`nation therapy consisting of interferon-α and
`ribavirin mainly acts by supporting host cell
`defence. These therapeutics are the prototypic
`representatives of indirect antiviral agents as they
`act on cellular targets. However, the therapy is not
`a cure, when considered from the long-term
`perspective, for almost half of the chronically
`infected patients. This draws attention to the
`urgent need for more efficient treatments. Novel
`anti-hepatitis C treatments under study are directed
`against a number of so-called direct antiviral
`targets such as polymerases and proteases, which
`are encoded by the virus. Although such direct
`antiviral approaches have proven to be successful
`in several viral indications, there is a risk of
`
`resistant viruses developing. In order to avoid resis-
`tance, the development of
`indirect antiviral
`compounds has to be intensified. These act on host
`cell targets either by boosting the immune
`response or by blocking the virus host cell interac-
`tion. A particularly interesting approach is the
`development of inhibitors that interfere with signal
`transduction, such as protein kinase inhibitors. The
`purpose of this review is to stress the importance of
`developing indirect antiviral agents that act on host
`cell targets. In doing so, a large source of potential
`targets and mechanisms can be exploited, thus
`increasing the likelihood of success. Ultimately,
`combination therapies consisting of drugs against
`direct and indirect viral targets will most probably
`provide the solution to fighting and eradicating
`hepatitis C virus in patients.
`
`Keywords: HCV, interferon, ribavirin, indirect
`(cellular) and direct (viral) antiviral targets,
`(in)direct antiviral agent/compound, protein kinase,
`signal transduction, glutathione peroxidase
`
`Introduction
`
`HCV: the disease and its consequences
`
`Hepatitis C is a serious threat to a significant percentage
`(1–2%) of the global population. This review summarizes
`the course of the disease and existing treatments, describes
`products under investigation and explains their mode of
`action on direct and indirect antiviral targets. Direct viral
`targets are those that are encoded by the viral genome.
`Indirect viral targets are those encoded by the host cell
`genome and are functions that are either usurped by the
`virus to allow its propagation or play important roles in the
`immune response and immunomodulation. We discuss
`here some exciting approaches to discovering and
`exploiting novel, cellular or indirect antiviral targets as
`potential therapies. These targets might be exploited to
`serve as a basis for the generation of new and powerful
`medications against hepatitis C.
`
`Chronic viral hepatitis is a common disease. More than 500
`million people suffer from chronic viral hepatitis world-
`wide, due to chronic infection with hepatitis B virus,
`hepatitis D virus or hepatitis C virus (HCV). Chronic viral
`hepatitis is the main cause of cirrhosis and hepatocellular
`carcinoma (HCC), which are responsible for major
`morbidity and mortality worldwide. There are 350 million
`cases of chronic hepatitis B infection and 170 million cases
`of chronic hepatitis C infection (Marcellin & Boyer, 2003).
`With no prophylactic or therapeutic vaccines available to
`date, this will create a major health crisis during the next
`decade. HCV escapes the immune system and establishes a
`persistent infection in approximately 75% of cases. These
`chronic carriers are at risk of developing life-threatening
`liver disease, such as cirrhosis and HCC. Five million
`
`©2005 International Medical Press 0956-3202
`
`69
`
`Gilead 2002
`I-MAK v. Gilead
`IPR2018-00211
`
`

`

`BM Klebl et al.
`
`people in Europe and 4 million people in the USA are
`chronically infected, with an estimated 20–50% of these
`patients likely to develop cirrhosis within the next 20–30
`years (Vogel, 2003). The entire spectrum of outcome ranges
`from a very slow progression over 50 years to an accelerated
`progression to cirrhosis within 5 years (Zoulim et al., 2003).
`In retrospective studies of post-transfusion hepatitis C,
`some 20–50% developed cirrhosis and 5–25% developed
`HCC after 10–30 years (Durantel et al., 2003) (Figure 1).
`Cases of decompensated liver cirrhosis ultimately undergo
`liver transplantation. Even after liver transplantation, the
`rate of relapses is high, which is most probably due to
`extrahepatic infection, subsequently resulting in re-
`infection of the transplanted liver. Several reports indicate
`that HCV can also infect organs and cell types other than
`the liver, particularly lymphoid cells (Dammacco et al.,
`2000). Furthermore, HCV RNA can persist at very low
`levels in the serum and peripheral lymphoid cells and can
`persist in peripheral blood monocytes for many years after
`spontaneous or antiviral therapy-induced resolution of
`chronic hepatitis C (Pham et al., 2004). Negative-strand
`HCV RNA has been reported in the brain, providing
`evidence that the central nervous system is an additional
`site of HCV replication (Forton et al., 2004). This may
`explain the cerebral dysfunction found in chronically
`
`HCV-infected patients. These extrahepatic infections
`might contribute to the immune-mediated pathogenesis of
`chronic liver disease and/or the development of auto-
`immune diseases, including mixed cryoglobulinaemia. The
`presence of abnormal serum levels of cryoglobulins can
`damage the kidneys and cause glomerulonephritis, a kidney
`disease affecting the capillaries of the glomeruli (the
`compact cluster of capillaries in the kidney that filter
`blood). The protein is characterized by oedema, raised
`blood pressure and excess protein in the urine.
`A prognosis for chronically infected HCV patients can
`be made, relying on biochemical and histological parame-
`ters (Marcellin & Boyer, 2003). Important parameters for
`the long-term prognosis of chronically infected patients are
`the initial level of viral load in the serum and the early viro-
`logical response towards interferon (IFN) treatment
`(Durantel et al., 2003). In general, ~25% of patients have
`normal serum alanine aminotransferase (ALT) levels
`despite detectable HCV RNA in serum; liver histological
`lesions are generally mild, cirrhosis is rare and their prog-
`nosis is good. The majority, ~50%, of chronically infected
`patients progress very slowly and the long-term risk of
`developing cirrhosis is low. Approximately 25% of the
`chronically infected patients have moderate to severe
`chronic hepatitis. Liver biopsies are used to diagnose
`
`Figure 1. Progression of HCV infection
`
`Responders
`(45–46%)
`
`Non-responders
`(35–54%)
`
`Treatment:
`PEG-IFNα/ribavirin
`
`Acute hepatitis C
`(45–46%)
`
`Chronic
`(75%)
`
`Cirrhosis
`(20–50%)
`
`Liver failure,
`HCC, transplant
`(25%)
`
`Resolved
`(25%)
`
`Stable disease
`(50–80%)
`
`Slowly progressive
`(75%)
`
`Flow chart for the progression and prognosis of acute hepatitis C infection. Patients suffering from acute hepatitis C have a 25% chance of
`resolving the disease and a 75% chance of developing chronic hepatitis C. Without treatment, chronic hepatitis C patients develop either
`stable disease or cirrhosis. Of cirrhotic patients, 75% are slowly progressive and 25% develop hepatocellular carcinoma (HCC), ultimately
`undergoing liver failure requiring transplantation. Of chronic patients, 46–65% who are treated with PEG-IFN-α/ribavirin combination are
`long-term responders. The remaining 35–54% are non-responders and subject to the same conditions as non-treated chronic patients.
`
`70
`
`©2005 International Medical Press
`
`

`

`moderate or severe chronic hepatitis. These patients are at
`elevated risk of developing HCV-related cirrhosis, which
`can lead to mortality due to portal hypertension, hepatic
`failure or HCC. These numbers are representative of non-
`treated HCV patients. Approximately 250000 chronically
`infected HCV patients receive therapy per year. Only
`46–65% of these patients respond to the current drug
`regimen. The remaining 35–54%, the so-called non-
`responders, face disease comparable with non-treated
`chronically infected patients,
`leading to a significant
`number of cirrhotic patients with an elevated risk of devel-
`oping HCC and liver failure (Figure 1).
`
`Incidences and transmission
`De novo infections are still considered to occur at the rate of
`20–25 cases per 100000 persons per year (Vogel, 2003).
`HCV is mainly transmitted through contact with blood
`and blood products, with blood transfusions and sharing of
`non-sterilized needles and syringes being the main routes
`(Poynard et al., 2003). With the advent of routine blood
`screening for HCV antibodies in 1991, transfusion-related
`hepatitis C has almost disappeared and the incidence has
`been in decline (Marcellin & Boyer, 2003).
`
`Treatments and perspectives
`Because of the small number of symptomatic patients,
`randomized trials to identify new regimens in acute
`hepatitis C have been rare. So far, the standard treatment
`for chronic HCV infection, IFN-α, has been found to be
`effective for acute hepatitis C (Poynard et al., 2003). The
`aim of the antiviral therapy is to cure viral infection and
`thereby prevent the progression of liver disease towards
`cirrhosis and HCC. Ten years ago, IFN-α monotherapy
`was approved by the Food and Drug Administration
`(FDA) as the basis of therapies for chronic viral hepatitis
`(Tan et al., 2002). Subsequently, a combination treatment
`(Davis et al., 1998; McHutchinson et al., 1998), consisting
`of IFN-α and ribavirin (Sidwell et al., 1972) was intro-
`duced 6 years ago. The IFN-α/ribavirin combination has
`been considerably improved by the introduction of pegy-
`lated interferons (PEG-IFNs) (Vogel, 2003). Using either
`PEG-IFN-α2b
`(PEG-Intron®;
`Schering-Plough,
`Kenilworth, NJ, USA) or PEG-IFN-α2a (Pegasys®;
`Hoffmann-LaRoche, Basel, Switzerland) in combination
`with ribavirin (Rebetol®; Schering-Plough or Copesus®;
`Hoffmann-LaRoche), sustained viral responses (SVRs) as
`high as 46–65% have been achieved for chronically
`infected patients (Figure 1). These clinically manifested
`results have turned the combination treatment into the
`most efficacious therapy for HCV currently available
`(Vogel, 2003).
`Only a minor fraction of chronically infected patients
`receive medication, indeed, less than 1%. The costs of
`
`Host cell targets in HCV therapy
`
`complications, including decompensated cirrhosis and liver
`transplantation, may far exceed the medication costs for
`PEG-IFN-α/ribavirin treatment. In the absence of a more
`affordable combination treatment, we expect a pharma-
`coeconomic health crisis for the industrialized world, let
`alone in the major areas of virus dissemination in Asia, with
`a total of 9 million people infected with HCV in the USA
`and Europe (Vogel, 2003). Since 35–54% of patients are
`still non-responders to the currently existing and expensive
`therapies, it has become obvious that there is a huge need
`for novel treatment alternatives and medications. Without
`effective treatment strategies, HCV-related morbidity and
`mortality are expected to increase nearly threefold by the
`year 2015. The age-adjusted death rate in 1999 was 1.8 per
`100000 persons in the USA (Kim, 2002). The develop-
`ment of small molecule drugs will become particularly
`important as they are typically associated with lower
`production and development costs. Novel treatment
`options should not only address the patient population of
`IFN non-responders, but also try to shorten the overall
`treatment period, which currently takes up to 24 months.
`Equally important, although difficult to achieve due to the
`quasispecies nature of HCV (a quasispecies is a family of
`closely related, but slightly different, viral genomes; viral
`genetic variants, derived from the original infecting virus,
`which are present during an infection) and the large
`number of HCV genotypes, is the development of preven-
`tive and therapeutic vaccines, which are currently being
`tested
`in Phase II clinical
`trials
`(Pawlotsky &
`McHutchison, 2004).
`
`HCV biology
`
`HCV has been classified as the sole member of a distinct
`genus, Hepacivirus, in the family Flaviviridae, which also
`includes flaviviruses and pestiviruses. Originally cloned in
`1989 (Choo et al., 1989), the viral genome is now well
`characterized. HCV is an enveloped particle harbouring a
`plus-strand RNA molecule that is ~9600 nucleotides in
`length (Bartenschlager & Lohmann, 2000; Bartenschlager,
`2002). The initiation of translation is mediated by the
`interplay of host and viral factors. An internal ribosome
`entry side (IRES), a complex RNA structure located at the
`5′ non-coding region, serves to bind directly to ribosomes
`to initiate protein synthesis (Durantel et al., 2003;
`Bartenschlager, 2002). The open reading frame (ORF)
`encodes a polyprotein of ~3000 amino acids in length,
`which is processed by both cellular and viral proteases into
`at least 10 discrete polypeptides (Figure 2A). The structural
`proteins (core or capsid, gpE1 and gpE2) are used for the
`assembly of new-progeny virus particles, whereas most of
`the non-structural (NS) proteins (p7, NS2, NS3, NS4A,
`NS4B, NS5A and NS5B) participate in the replication of
`
`Antiviral Chemistry & Chemotherapy 16:2
`
`71
`
`

`

`BM Klebl et al.
`
`Figure 2. Schematic representation of (A) the HCV genome and (B) the sub-genomic replicon
`
`A
`
`5‘
`
`NTR
`IRES
`
`Structural proteins
`
`Non-structural proteins
`
`C
`
`E1
`
`E2
`
`p7
`
`2
`
`3
`
`4A 4B
`
`5A
`
`5B
`
`NTR
`
`3‘
`
`Translation and processing
`
`Signal peptidase
`
`NS2–NS3
`proteinase
`
`NS3 proteinase
`
`C
`
`E1
`
`E2
`
`p7
`
`2
`
`3
`
`4A
`
`4B
`
`5A
`
`5B
`
`Core
`
`Envelope
`
`Proteinase
`Helicase
`
`Phospho-
`protein
`
`RNA-dependent
`RNA polymerase
`
`B
`
`5‘
`
`NTR
`
`neo
`
`EMCV
`
`IRES
`
`Non-structural proteins
`
`3
`
`4A 4B
`
`5A
`
`5B
`
`NTR
`
`3‘
`
`(A) The major structural proteins include the core (C) and the envelope proteins (E1 and E2). The non-structural proteins p7, NS2, NS3,
`NS4A/NS4B and NS5A/NS5B are indicated. At the 5′ non-translated region (NTR) resides the internal ribosomal entry site (IRES), which is high-
`ly conserved and represents a site for development of translation inhibitors such as antisense oligonucleotides, ribozymes and small intefer-
`ing RNAs (siRNAs). NS3 encodes a protein with a specific protease and helicase activity. The NS5A region encodes for a phosphoprotein and
`the NS5B for an RNA-dependent RNA polymerase enzyme, both important for viral replication. They also represent sites for the development
`of specific viral enzyme inhibitors. Other potential enzyme targets include the HCV-specific proteases (NS2/3 and NS3). These enzymes are
`involved in processing the viral polyprotein at specific sites as indicated. (B) The subgenomic replicon has been generated by replacing the
`region that encodes the core protein up to the NS2-encoding region by the neomycin phosphotransferase gene (neo) and the IRES of the
`encephalomyocarditis virus (EMCV). This IRES drives the translation of the HCV polypeptide from NS3 to NS5B, whereas the selectable marker
`neo is expressed under the control of the original HCV IRES of the 5′ non-translated region.
`
`the viral genome. During viral replication, the viral genome
`acts as a template for the synthesis of negative-strand
`RNA, which, in turn, is a template for the production of
`excess amounts of positive-strand RNA progeny. Details
`concerning the initiation of the synthesis of the positive-
`strand RNA are not clearly known and only sparse infor-
`mation is available in terms of initial and late phases, for
`example, entry and morphogenesis of viral particles, since
`the field lacks a reproducible and efficient cell culture
`system for viral replication (Bartenschlager & Lohmann,
`2000; Bartenschlager, 2002; Dymock et al., 2000).
`
`HCV consists of six different genotypes (genotypes 1–6).
`Knowledge of the genotype or serotype is helpful for predic-
`tion of SVR and the choice of treatment duration. Genotypes
`do not change during the course of an infection. Response
`rates to treatment with the combination of PEG-IFN and
`ribavirin are 88% for genotypes 2 and 3, and 48% for geno-
`types 1, 4, 5 and 6 (Poynard et al., 2003). Unfortunately,
`genotype 1 is the most frequent genotype in Europe and the
`USA and is present in 60–80% of cases (Marcellin & Boyer,
`2003). These insufficient response rates underscore the need
`for the development of novel and efficient genotype-
`
`72
`
`©2005 International Medical Press
`
`

`

`independent treatments and also argue for the further
`improvement of existing cellular and animal models.
`
`Tissue culture model: the replicon system
`
`Many aspects of HCV disease can only be addressed in
`vivo, for example, the influence of the immune system. For
`many investigatiors, however, it is sufficient and desirable
`to work with a less complex, well-controlled system in
`vitro. The recently generated HCV-replicon system, in
`which expression of the HCV non-structural proteins
`drives the replication of a subgenomic HCV RNA, fulfills
`these criteria (for reviews, see Bartenschlager & Lohmann,
`2001; Bartenschlager, 2002).
`The human hepatoma cell line (Huh-7) was trans-
`fected with subgenomic HCV RNAs called replicons
`(Lohmann et al., 1999). These were derived from a cloned
`full-length HCV genome of genotype 1b (Figure 2A) by
`replacing the reading frame for the N-terminal proteins
`(including p7 or NS2) with the neomycin phosphotrans-
`ferase gene (neo) downstream of the HCV IRES.
`Translation of the HCV NS2-5B or NS3-5B region was
`directed by the IRES of the encephalomyocarditis virus
`(EMCV) inserted downstream of the neo gene (Figure
`2B). Upon transfection of Huh-7 cells, only those in
`which HCV RNA replication occurs develop continuous
`resistance against the drug geniticin due to neo expression.
`Cell lines obtained from such resistant colonies contain
`high
`levels of replicon RNAs and viral proteins
`(Lohmann et al., 1999; Pietschmann et al., 2001). This
`allows the study of HCV RNA replication as well as of
`translation and processing of those HCV proteins present
`in the system (Figure 2).
`The development of subgenomic replicons containing
`HCV non-structural proteins that replicate in human
`hepatoma cells, now provides a system to test or screen
`candidate drugs that target the non-structural gene prod-
`ucts. Thus, the antiviral efficacy of BILN 2061, a NS3
`protease inhibitor, the development of which was put on
`hold after entering Phase II clinical trials (see Table 1), was
`determined in the HCV replicon cell culture system. BILN
`2061 has an IC50 of 4 nM (genotype 1a) and 3 nM (geno-
`type 1b), while cytotoxicity analysis in parental Huh-7 cells
`produced a cytotoxic concentration (CC50) of 16–35 μM
`(Pause et al., 2003; Lamarre et al., 2003). As these sub-
`genomic replicons do not produce infectious virions, the
`option of studying encapsidation or cell-to-cell viral trans-
`mission is not provided. Furthermore, the replicon cell lines
`are of clonal origin and have, apart from adaptive muta-
`tions,
`identical HCV sequences (Krieger et al., 2001).
`However, the genetic heterogeneity of HCV suggests that
`future drugs should display activity against a broad range of
`HCV genotypes, subtypes and quasispecies.
`
`Host cell targets in HCV therapy
`
`In summary, the HCV-replicon RNA replicates to fairly
`high levels in Huh-7 cells and provides, for the first time, a
`genetic system to study HCV RNA replication and a cell-
`based assay screen for HCV inhibitors. Such a replicon-
`based screen for anti-HCV substances has been described in
`a recent study on the nucleoside antimetabolite-mediated
`reduction of HCV RNA (Stuyver et al., 2003). In this study,
`a specific anti-HCV replicon effect was defined as minimal
`interference with the exponential cell growth, minimal
`reduction in cellular host RNA levels and reduction of the
`HCV RNA copy number per cell compared with that of the
`untreated control (Stuyver et al., 2003).
`
`Animal models
`
`Developing robust animal model systems for HCV is highly
`desirable and some progress has recently been achieved
`(reviewed by Pietschmann & Bartenschlager, 2003). Besides
`the fact that ethical considerations do not permit experi-
`ments with humans, an animal system may allow for moni-
`toring of the entire cycle of viral replication, from infection
`of naive tissues to full-blown viraemia. Since acute HCV
`infection often happens without any obvious symptoms,
`these patients do not consult a physician. Thus, valuable
`information about early events of HCV infection and
`remission is lacking. A model system will allow the study of
`each phase of the disease under controlled conditions.
`Establishing a model system is hampered by the fact that
`HCV infects only humans and chimpanzees, primarily
`targeting hepatocytes. The determinants of the restricted
`host and tissue specificity are not understood. Despite the
`extremely robust replication rate of HCV in humans, efforts
`to propagate this virus in cell culture have been frustratingly
`unsuccessful. Therefore, the use of surrogate virus models
`closely related to HCV, such as the bovine viral diarrhoea
`virus (BVDV) and the tamarin GB virus-B (GBV-B), both
`which belong
`to Flaviviridae, provides alternative
`approaches. GBV-B is most closely related to HCV and is,
`therefore, a good surrogate model. For instance, one poten-
`tial mechanism of action of the pleiotropic antiviral agent,
`ribavirin, was verified using the GBV-B model. The
`antiviral effect of ribavirin does not seem to be solely based
`on the inhibition of inosine 5′-monophosphate dehydroge-
`nase (IMPDH) and reduction of intracellular pools of GTP
`and dGTP, but also by the incorporation of ribavirin
`triphosphate into viral RNA and induction of error-prone
`replication (Lanford et al., 2001). However, the biological
`activity of a tested compound in these systems does not
`necessarily translate into efficacy for human HCV infection.
`
`Transplant mouse models
`Mice are the preferred models for scientific studies for a
`number of reasons (for example, short breeding cycles,
`
`Antiviral Chemistry & Chemotherapy 16:2
`
`73
`
`

`

`BM Klebl et al.
`
`Table 1. Current HCV development pipeline
`
`Phase I
`
`Phase II
`
`Phase III
`
`Phase IV
`
`New interferons
`Albuferon* (Human Genome Sciences)
`
`PEG alphacon* (InterMune)
`Transfersome containing IFN-α* (IDEA)
`
`Other biologicals (antisense, vaccines, etc)
`HCV/MF59† (Chiron)
`
`Immunomodulators
`
`Oral IFN-α*
`(Amarillo Biosciences)
`IFN-γ1b* (InterMune)
`Omega Interferon* (BioMedicine)
`Multiferon* (Viragen)
`
`ISIS 14803† (ISIS Pharmaceuticals/Elan)
`E-1† (Innogenetics)
`Civacir† (NABI)
`HepeX™-C† (XTL)
`Rituximab† (Rituxam)
`(Genentech/IDEC)
`HCV vaccine† (Intercell)
`
`Merimebodib-VX-497‡ (Vertex)
`Ceplene‡ (Maxim)
`ANA245‡ (Anadys)
`Viramidine‡,§ (Valeant Pharmaceuticals)
`CPG 10101‡ (Coley
`Pharmaceuticals)
`
`REBIF*
`(Ares-Serono)
`
`Infergen*
`(InterMune)
`
`Zadaxin‡ (SciClone)
`
`Direct antiviral small molecules
`HCV-086 (ViroPharma/Wyeth)
`VX-950¶ (Vertex)
`SCH-7¶ (Schering)
`R803§,£ (Rigel Pharmaceuticals)
`JTK-109§ (AKROS Pharma/Japan Tobacco)
`R1479§ (Roche)
`
`Antifibrotics
`
`NM283§ (Idenix Pharmaceuticals)
`BILN 2061¶,r (Boehringer Ingelheim)
`JTK-003§ (AKROS Pharma/Japan Tobacco)
`KPE02003002§ (Kemin Pharma)
`UT-231-B$ (United Therapeutics)
`
`Amantadine$
`(Endo Labs Solvay)
`
`IDN-6556♠(Idun Pharmaceuticals)
`
`IP-501u (Indevus)
`
`*Interferon family, covering long-lasting interferons such as albuferon, which is a fusion of human interferon and albumin, and multiferon.
`REBIF is IFN-β1a and omega interferon is a new formulation intended to target the liver specifically. IFN-γ1b is aimed at treating liver fibrosis.
`IFN-α is administered in low doses, which get absorbed through mucosal membranes (oral IFN-α). Infergen® is a consensus interferon, which
`was generated through comparing the amino acid sequences of the then-known subtypes of INF-α and assigning the most commonly occurring
`amino acid sequence to a new protein, thereby creating a ‘consensus’ interferon.
`†Biologicals, such as vaccines (HCV/MF59, E-1), antibodies (Rituximab, Civacir) and antisense (ISIS 14803).
`‡Immunomodulator, boosting the immune system, like the IMPDH inhibitor VX-497 (merimebodib) and viramidine, the toll-like receptor 7
`agonist ANA245 (isatoribine), the toll-like receptor 9 agonist CPG 10101, histamine (Ceplene) and thymosin α-1 (Zadaxin).
`§Small molecule nucleosidic and non-nucleosidic polymerase inhibitors (JTK-003, JTK-109, R803, R1479, NM283, viramidine). Viramidine pre-
`sents a special case, since it is a prodrug of ribavirin, which is specifically targeted towards liver cells. The molecular target for the antiviral
`compound KPE02003002 has not been revealed to our knowledge.
`¶Small molecule inhibitors of the viral protease (BILN2061, SCH-7, VX-950).
`$Small molecule inhibitors of the viral p7 ion channel. UT-231-B belongs to the iminosugar derivatives, for which activity on p7 has been
`demonstrated. Amantadine is a pleiotropic agent that has been launched for the treatment of influenza. It blocks the M2 ion channel of the
`influenza A virus and prevents the passage of H+ ions (DeClercq, 2004). Therefore, it seems likely that amantadine might act via inhibiting the
`p7 ion channel.
`♠IDN-6556 is a caspase inhibitor, which is believed to preserve cell structure and protect liver from damage caused by HCV.
`uPurified phospholipid.
`rPhase II trials have been halted and further development of this drug is reportedly suspended owing to cardiac toxicity in non-human pri-
`mates at a supra-efficacious dose.
`£Rigel have just announced insignificant clinical effects for R803 in a Phase I/II HCV trial due to poor bioavailability
`(http://www.rigel.com/rigel/corporate).
`Underlined text represents indirect antiviral targets or host cell targets, which are modulated through the treatment. Italic text represents
`direct antiviral targets. There are two exceptions: viramidine since it is a modified version of ribavirin. Ribavirin is a pleiotropic agent, exert-
`ing direct and indirect antiviral effects. The same is true for amantadine, since amantadine does not only target the virally encoded M2 pro-
`ton channel (Lear, 2003). Amantadine is an antagonist of the N-methyl D-aspartate receptor as well (Magnet et al., 2004).
`
`74
`
`©2005 International Medical Press
`
`

`

`simple handling and genetic characterization). Mice cannot
`be infected directly with HCV due to the restricted host
`range of the virus. Therefore, transgenic mice expressing
`one or several viral proteins were created to analyse their
`effects on liver pathology, but their meaningfulness for
`drug development remains somewhat unclear.
`Significantly more progress has been reported on the
`generation of transplant mouse models. These transplant
`mouse models might lead to the development of more suit-
`able small animal models in the future. Propagation of
`HCV in these chimeric mouse models has recently been
`achieved (Mercer et al., 2001; Ilan et al., 2002).
`The principle of the first model is that immunocompro-
`mised mice, unable to develop murine hepatocytes due to a
`transgene, were engrafted with human hepatocytes isolated
`from fresh livers and shown to be susceptible to HCV infec-
`tion and replication. To achieve this system, Mercer and
`colleagues transplanted human hepatocytes into immunod-
`eficient transgenic mice (SCID, homozygous for the SCID
`trait) carrying the urokinase-type plasminogen-activator
`gene controlled by the albumin promoter (Alb-uPA mice).
`The Alb-uPA transgenic mouse, developed in 1990 to study
`neonatal bleeding disorders, carries a tandem array of four
`murine urokinase genes controlled by an albumin promoter.
`This transgene targets urokinase over-production to the
`liver resulting in a profoundly hypofibrinogaenemic state
`and accelerated hepatocyte death. Normal human hepato-
`cytes were transplanted into these SCID mice carrying the
`plasminogen activator transgene Alb-uPA, thus generating
`mice with chimeric human livers. After inoculation with
`infected human serum, viral infection persisted beyond 4–5
`weeks only in homozygous Alb-uPA mice. The shorter
`persistence of infection in Alb-uPA heterozygous mice is
`attributed to a much lower survival of transplanted human
`hepatocytes in these mice relative to homozygous mice.
`Approximately 75% of homozygous Alb-uPA mice inocu-
`lated with serum from hepatitis C patients developed HCV
`titres. The viral titres reached 3×104–3×106 copies/ml in the
`blood of infected mice and were equal to or higher than
`those present in patients with chronic hepatitis C. This
`novel mouse model supports prolonged HCV infection for
`15–17 weeks, and even for 35 weeks in one mouse. Infection
`could be serially passaged through three generations of
`mice, confirming both synthesis and release of infectious
`viral particles. Thus, this is the first murine model that may
`be suitable for studying human HCV in vivo (Mercer et al.,
`2001). However, this system is laborious and requires special
`expertise to isolate and transplant human hepatocytes and
`to maintain a colony of fragile immunodeficient mice with
`an approximately 35% mortality in newborns due to a defect
`in blood coagulation. This may be the reason why, 3 years
`after its introduction into the scientific community, this
`system has not reached a broad application. There is also a
`
`Host cell targets in HCV therapy
`
`scientific obstacle: in humans with chronic hepatitis C, the
`injury of hepatocytes is not directly caused by HCV infec-
`tion but rather is a consequence of the destruction of
`infected hepatocytes by cytotoxic lymphocytes. A crucial
`question is whether these mice will develop liver disease
`confined to the transplanted human hepatocytes or whether
`the immunosuppressed mice will be infected but free of
`disease.
`In another approach, immunodeficient BNX mice were
`irradiated with a lethal dose, rescued by transplantation of
`bone marrow cells originating from severe combined
`immunodeficient SCID mice and finally, had liver frag-
`ments from ex vivo HCV-infected humans transplanted
`under the kidney capsule (Galun et al., 1995). The resulting
`system, made up from three genetically disparate sources of
`tissue, was termed ‘Trimera’. Viraemia (positive-strand
`HCV-RNA levels) in HCV-Trimera mice peaked at
`approximately day 18 after liver transplantation, and viral
`replication in liver grafts was evidenced by the presence of
`specific negative-strand HCV RNA. Recently,
`it was
`claimed that up to 85% of the transplanted animals of this
`Trimera-mouse system were HCV-infected and that an
`antibody was effective in reducing the viraemia in this
`model (Ilan et al., 2002). This anti-HCV monoclonal anti-
`body (MAb) HCV ABXTL68 was developed from the
`peripheral blood lymphocytes of an HCV-positive patient.
`It was characterized as a fully human high-affinity IgG1
`subtype MAb against the HCV E2 envelope protein. The
`antibody was further developed and is currently being
`studied in clinical trials for chronic HCV patients
`(HepeX™-C in Table 1). So far, the antibodies have been
`shown to be safe, tolerable and could significantly reduce
`viral load (Ilan et al., 2002; Dagan & Eren, 2003).
`Another antiviral drug, I70,
`inhibiting HCV RNA
`translation, was also effective in reducing the viraemia in
`this model. The I70 molecule was selected via high-
`throughput screening in a cell-based IRES assay designed
`for screening of HCV IRES inhibitors (Ilan et al., 2002).
`In conclusion, both systems should help to expedite
`anti-HCV drug evaluation if available to the pharmaceu-
`tical industry without extreme costs and legal complica-
`tions, but they also have their limitations, especially in light
`of the role of host cell targets, which play an important role
`in the immune response and the HCV replication process.
`
`Chimpanzee model
`Until recently the only well-established animal model
`supporting HCV replication was the chimpanzee model.
`The successful infection of this animal was invaluable for
`the initial characterization of HCV, which finally led to the
`identification of infectious cDNA clones of the virus (Choo
`et al., 1989). The clinical course of virus infection observed
`in chimpanzee and humans shows some similarities. Apart
`
`Antiviral Chemistry & Chemotherapy 16:2
`
`75
`
`

`

`BM Klebl et al.
`
`from humans,

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