throbber
Downloaded from
`
`http://jvi.asm.org/
`
` on March 22, 2018 by guest
`
`JOURNAL OF VIROLOGY, Mar. 2007, p. 3005–3008
`0022-538X/07/$08.00⫹0 doi:10.1128/JVI.02083-06
`Copyright © 2007, American Society for Microbiology. All Rights Reserved.
`
`Vol. 81, No. 6
`
`Synergy of Small Molecular Inhibitors of Hepatitis C Virus Replication
`Directed at Multiple Viral Targets䌤
`David L. Wyles,1* Kelly A. Kaihara,1 Florin Vaida,2 and Robert T. Schooley1
`Department of Medicine, Division of Infectious Diseases,1 and Department of Family and Preventive Medicine,2
`University of California, San Diego, La Jolla, California
`
`Received 22 September 2006/Accepted 10 December 2006
`
`Chronic hepatitis C virus (HCV) infection is a significant worldwide health problem with limited therapeutic
`options. A number of novel, small molecular inhibitors of HCV replication are now entering early clinical trials
`in humans. Resistance to small molecular inhibitors is likely to be a significant hurdle to their use in patients.
`A systematic assessment of combinations of interferon and/or novel anti-hepatitis C virus agents from several
`different mechanistic classes was performed in vitro. Combinations of inhibitors with different mechanisms of
`action consistently demonstrated more synergy than did compounds with similar mechanisms of action. These
`results suggest that combinations of inhibitors with different mechanisms of action should be prioritized for
`assessment in clinical trials for chronic hepatitis C virus infection.
`
`Chronic hepatitis C virus (HCV) infection is a major world-
`wide health problem; in the United States, an estimated 3
`million persons are chronically infected (4). Estimates of the
`health care burden of chronic HCV infection predict a drastic
`increase in hospitalizations and medical costs related to com-
`plications such as cirrhosis and hepatocellular carcinoma over
`the next 1 to 2 decades (3). Effective and better-tolerated
`therapy for HCV could effectively stem this tide (7).
`Current interferon-based therapy for chronic HCV infection
`results in sustained responses in roughly 55% of patients and is
`accompanied by significant toxicity. Genotype 1 HCV, the
`most prevalent genotype in the United States, responds less
`well to therapy with pegylated interferon plus ribavirin, with
`response rates of 42 to 46% (11, 23). These limitations have
`spurred an intense drug discovery effort, resulting in a number
`of promising compounds (8).
`Hepatitis C virus replication takes place in the cytoplasm,
`with the replication complex being tightly associated with lipid
`membranes (1). Key components of the replication complex
`include several promising antiviral
`targets,
`including the
`NS3/4A protease and the NS5B RNA-dependent RNA poly-
`merase. A number of candidate protease inhibitors (PIs) which
`have excellent potency in vitro have been developed (2, 17, 20);
`several of these compounds have also been evaluated in phase
`I/II trials, with encouraging results (15, 16, 29, 36). Resistance
`to this class of inhibitors has been described, with some muta-
`tions conferring cross-resistance to several compounds (17, 18,
`21, 34, 35).
`The NS5B RNA polymerase is also essential for viral repli-
`cation, and a number of nucleoside inhibitors and nonnucleo-
`side inhibitors (NNIs) of the HCV polymerase with potent
`activity in vitro and in early clinical trials have been described
`(5, 12, 13, 27, 30). Resistance to both nucleoside and non-
`nucleoside inhibitors in vitro has been described (22, 24, 26).
`
`* Corresponding author. Mailing address: 9500 Gilman Drive, MC
`0711, La Jolla, CA 92093. Phone: (858) 822-1779. Fax: (858) 822-5362.
`E-mail: dwyles@ucsd.edu.
`䌤 Published ahead of print on 20 December 2006.
`
`We have assessed a number of combinations of HCV inhibitors
`with several molecular targets currently in development, using
`an HCV genotype 1 replicon-based luciferase reporter system.
`Replicon constructs. The BM4-5 replicon is a subgenomic
`HCV genotype 1b replicon which contains a deletion of a
`serine in NS5A and has been previously described (14). The
`firefly luciferase gene was inserted in the BM4-5 replicon, in a
`manner previously described (33), to create a luciferase/neo-
`mycin phosphotransferase fusion protein (FEO) and the rep-
`licon (BM4-5 FEO). Briefly, the Photinus pyralis luciferase
`gene was amplified using primers coding for the AscI restric-
`tion site. Following amplification, both the BM4-5 plasmid and
`luciferase PCR product were restriction digested with AscI.
`Ligation was then carried out to insert the luciferase gene in
`phase with the neomycin phosphotransferase gene, creating
`the desired BM4-5 FEO replicon. The sequence of the repli-
`con was verified by DNA sequencing.
`Cell culture. Human hepatoma Huh-7.5.1 cells (a kind gift
`from Francis Chisari, Scripps Research Institute, La Jolla, CA)
`and BM4-5 FEO cells stably expressing the BM4-5 FEO rep-
`licon were grown at 37°C and 5% CO2 in Dulbecco’s modifi-
`cation of Eagle’s medium supplemented with 2 mM L-glu-
`tamine, 100 units/ml penicillin, 100 ␮g/ml streptomycin, and
`10% fetal bovine serum. BM4-5 FEO cells were additionally
`grown in the presence of 500 ␮g/ml of G-418.
`Transfection and clone selection. The BM4-5 FEO plasmid
`was linearized with ScaI. In vitro transcription (Megascript;
`Ambion) was carried out according to the manufacturer’s in-
`struction to yield BM4-5 FEO RNA. Transfection was per-
`formed as previously described (32). Four hundred microliters
`of a Huh-7.5.1 cell suspension (107 cells/ml) was placed in a
`0.4-cm cuvette with 10 ␮g of BM4-5 FEO RNA. The mixture
`was electroporated (Bio-Rad Gene Pulser) at 270 V and 975
`␮F and transferred to a 10-cm tissue culture dish. G-418 at 500
`␮g/ml was added at 24 h, and the medium was changed every
`3 to 4 days. Individual G-418-resistant colonies were visible
`within 2 to 3 weeks. Individual colonies were harvested and
`expanded for characterization of luciferase expression.
`Gilead 2003
`I-MAK v. Gilead
`IPR2018-00211
`
`3005
`
`

`

`3006
`
`NOTES
`
`J. VIROL.
`
`TABLE 1. Activities of different small molecular inhibitors in the
`BM4-5 replicon
`
`Compound
`
`Structure
`
`HCV BM4-5
`replicon
`IC50 (nM)a
`
`Downloaded from
`
`http://jvi.asm.org/
`
` on March 22, 2018 by guest
`
`a The IC50 is the average ⫾ standard error of mean of the results from at least
`three independent experiments.
`
`The IC50 for each of the individual compounds is listed in
`Table 1. CI50, CI70, and CI90 refer to the combination index at
`the IC50, IC70, and IC90, respectively, of each drug. All com-
`pounds tested were additive (CI50 and CI70) or mildly syner-
`gistic (CI90) with alpha interferon. Antagonism was not dem-
`onstrated for any combination of small molecular inhibitors,
`including compounds targeting the same viral protein. Signif-
`icantly more synergy was demonstrated between compounds in
`the group combining two small molecular inhibitors targeting
`the same viral enzyme (in this case NS3 protease) than be-
`tween the group of compounds combined with alpha interferon
`at both the CI70 (P ⫽ 0.043) and CI90 (P ⫽ 0.017) levels. There
`was no significant difference between the groups at the CI50
`level (P ⫽ 0.108) (Fig. 1). Similarly, the group consisting of two
`inhibitors with different viral targets showed significantly lower
`combination indices than either of the other two groups, i.e.,
`compounds with interferon (P ⬍ 0.001 at all levels) or com-
`pounds with same mechanism of action (P ⫽ 0.038 and 0.037
`at the CI50 and CI70 levels, respectively). The comparison of
`CI90s between small molecular inhibitors with the same and
`different viral targets showed a trend toward a lower combina-
`tion index in the group with two compounds with different viral
`targets (P ⫽ 0.056 at the CI90 level) (Fig. 1). None of the
`compounds or combinations showed cytotoxicity at the con-
`centrations tested in the activity and synergy studies (data not
`shown).
`Small molecular inhibitors of the HCV protease and poly-
`
`Luciferase compound assay. BM4-5 FEO cells were seeded
`into 96-well plates at a density of 10,000 cells per well in 100 ␮l
`medium. After allowing 4 h for attachment, compounds were
`added to wells at the specified concentrations. All conditions
`were run in triplicate. Cells and compounds were incubated for
`48 h. The luciferase assay (Bright-Glo; Promega) was carried
`out according to the manufacturer’s instructions. Luciferase
`activity was determined using a microplate luminometer (Ve-
`ritas microplate luminometer; Turner Biosystems). The rela-
`tive light units (RLU) for each condition were reported as the
`mean ⫾ the standard error of the mean for the three wells.
`Compounds tested. Compounds tested included two pep-
`tidomimetic HCV PIs, BILN 2061 (16) and a Vertex PI (19)
`(Vicki Sato, Vertex Pharmaceuticals, Cambridge, MA); a
`GlaxoSmithKline trans-lactam PI active-site mimic (2) (Karen
`Romines, GlaxoSmithKline, Research Triangle Park, NC); one
`nucleoside analog HCV RNA-dependent RNA polymerase
`inhibitor (RdRpI), 2⬘-C-methyladenosine (10) (William Lee,
`Gilead Sciences, Foster City, CA); one nonnucleoside GSK
`benzo-thiadiazine RNA polymerase inhibitor directed at the
`“thumb” region of the polymerase (Karen Romines, Glaxo-
`SmithKline) (9); and alpha interferon (Interferon-␣A; Sigma-
`Aldrich).
`The 50% inhibitory concentration (IC50) of each compound
`was determined independently and used to set the range of
`concentrations used for the synergy experiments. Each com-
`pound was tested singly and in combination at two twofold
`serial dilutions above and below the IC50. The ratio of the two
`compounds tested remained fixed across the dosing range.
`Potential cytotoxicity of individual compounds and all combi-
`nations was assessed using a luminescent ATP-based cell via-
`bility assay (Cell Titer-Glo; Promega).
`Data analysis. Determinations of compound interactions
`were based on the median-effect principle and the multiple
`drug effect equation as described by Chou and Talalay (6).
`Combination indices (CIs) were determined using Calcusyn
`(Biosoft) for each experiment at the IC50, IC70, and IC90 levels.
`In total, 15 combinations were evaluated with from three to
`five replicates per condition; this yielded a total of 61 data
`points per CI level analyzed. A CI of ⬍0.9 was considered
`synergistic, a CI of ⱖ0.9 or ⱕ1.1 was considered additive, and
`a CI of ⬎1.1 was deemed antagonistic.
`Statistical analysis. At each of the three inhibitory concen-
`trations evaluated (IC50, IC70, and IC90) the CIs in the three
`synergy groups were compared using a linear mixed-effects
`model allowing for different means in the three synergy groups
`and random effects for the individual drug combinations. The
`random effects were not significant (likelihood ratio test), in-
`dicating no statistical difference in CI values between the an-
`tiviral compound combinations in the same synergy group. The
`CI replicates were further compared between synergy groups
`by using the Wilcoxon rank test.
`Synergy of small molecular inhibitors. Transfection of Huh-
`7.5.1 cells with BM4-5 FEO RNA yielded numerous (⬎50)
`G-418-resistant clones. Individual clones were expanded and
`assessed using the luciferase assay to determine the individual
`clones with highest RLU per cell. Four clones yielded from
`30,000 to 50,000 RLU per 10,000 cells at 48 h (data not
`shown); these clones were expanded and used for all subse-
`quent studies.
`
`

`

`VOL. 81, 2007
`
`NOTES
`
`3007
`
`Downloaded from
`
`http://jvi.asm.org/
`
` on March 22, 2018 by guest
`
`FIG. 1. CI50s, CI70s, and CI90s for the compound combinations evaluated. Dotted lines at combination index values of 0.9 and 1.1 indicate the
`boundaries of an additive interaction. The P values displayed (ⴱ) are for analyses at the CI70 level.
`
`merase show antiviral activity in our genotype 1 replicon sys-
`tem. Most importantly, no combination of small molecular
`inhibitors of HCV replication demonstrated antagonism in our
`system, including those with the same mechanism of action or
`viral target. Combinations of inhibitors targeting different viral
`proteins (PI-RdRpI or PI-NNI) or with different mechanisms
`of
`inhibiting the same viral protein (RdRpI-NNI) were
`strongly synergistic and had significantly lower combination
`indices than the other two groups. Combinations targeting the
`same site within a viral protein showed lesser degrees of syn-
`ergy or were additive, but they still possessed significantly
`lower combination indices than the group composed of the
`same compounds with alpha interferon. It is important to re-
`member that the definition of synergy as a CI of less than 0.9
`is an arbitrary distinction (along a continuum) and thus does
`not preclude two inhibitors which occupy the same site from
`being “synergistic” according to a CI of ⬍0.9. Additionally,
`metabolic interactions between compounds or the impact of
`divergent resistance pathways on different compounds may
`also affect the appearance of drug-drug interactions as assessed
`by the combination index.
`HCV, like human immunodeficiency virus type 1, possesses
`an error-prone RNA polymerase, and it replicates to levels 10-
`to 100-fold higher than those of human immunodeficiency
`virus type 1 in chronically infected individuals (25, 28). These
`characteristics suggest that selection of drug-resistant viral
`variants will be a challenge to the use of small molecular
`inhibitors. In fact, resistance to these compounds both in vitro
`and in vivo has already been described (17, 24, 26, 31, 34).
`Synergistic combinations of HCV inhibitors may produce
`
`greater viral load decreases in vivo and could potentially delay
`the appearance of multiply drug-resistant virus. This system
`provides a useful approach for the in vitro testing of antiviral
`combinations in anticipation of rationally designed clinical
`studies of combination chemotherapy directed at HCV. Our
`results support the evaluation of combinations of small molec-
`ular inhibitors in human clinical trials and further suggest that
`combinations with different mechanisms of action may be par-
`ticularly attractive.
`
`This work was funded in part by a 2005 developmental grant from
`the UC San Diego Center for AIDS Research, an NIH-funded pro-
`gram (no. 5P30 AI-36214).
`
`REFERENCES
`1. Aizaki, H., K. J. Lee, V. M. Sung, H. Ishiko, and M. M. Lai. 2004. Charac-
`terization of the hepatitis C virus RNA replication complex associated with
`lipid rafts. Virology 324:450–461.
`2. Andrews, D. M., M. C. Barnes, M. D. Dowle, S. L. Hind, M. R. Johnson, P. S.
`Jones, G. Mills, A. Patikis, T. J. Pateman, T. J. Redfern, J. E. Robinson,
`M. J. Slater, and N. Trivedi. 2003. Pyrrolidine-5,5-trans-lactams. 5. Pharma-
`cokinetic optimization of inhibitors of hepatitis C virus NS3/4A protease.
`Org. Lett. 5:4631–4634.
`3. Armstrong, G. L., M. J. Alter, G. M. McQuillan, and H. S. Margolis. 2000.
`The past incidence of hepatitis C virus infection: implications for the future
`burden of chronic liver disease in the United States. Hepatology 31:777–782.
`4. Armstrong, G. L., A. Wasley, E. P. Simard, G. M. McQuillan, W. L. Kuhnert,
`and M. J. Alter. 2006. The prevalence of hepatitis C virus infection in the
`United States, 1999 through 2002. Ann. Intern. Med. 144:705–714.
`5. Chan, L., T. J. Reddy, M. Proulx, S. K. Das, O. Pereira, W. Wang, A.
`Siddiqui, C. G. Yannopoulos, C. Poisson, N. Turcotte, A. Drouin, M. H.
`aoui-Ismaili, R. Bethell, M. Hamel, L. L’Heureux, D. Bilimoria, and N.
`Nguyen-Ba. 2003. Identification of N,N-disubstituted phenylalanines as a
`novel class of inhibitors of hepatitis C NS5B polymerase. J. Med. Chem.
`46:1283–1285.
`6. Chou, T. C., and P. Talalay. 1984. Quantitative analysis of dose-effect rela-
`
`

`

`Downloaded from
`
`http://jvi.asm.org/
`
` on March 22, 2018 by guest
`
`3008
`
`NOTES
`
`J. VIROL.
`
`tionships: the combined effects of multiple drugs or enzyme inhibitors. Adv.
`Enzyme Regul. 22:27–55.
`7. Davis, G. L., J. E. Albright, S. F. Cook, and D. M. Rosenberg. 2003. Pro-
`jecting future complications of chronic hepatitis C in the United States. Liver
`Transpl. 9:331–338.
`8. De Francesco. R., L. Tomei, S. Altamura, V. Summa, and G. Migliaccio.
`2003. Approaching a new era for hepatitis C virus therapy: inhibitors of the
`NS3-4A serine protease and the NS5B RNA-dependent RNA polymerase.
`Antiviral Res. 58:1–16.
`9. Dhanak, D., K. J. Duffy, V. K. Johnston, J. Lin-Goerke, M. Darcy, A. N.
`Shaw, B. Gu, C. Silverman, A. T. Gates, M. R. Nonnemacher, D. L. Earnshaw,
`D. J. Casper, A. Kaura, A. Baker, C. Greenwood, L. L. Gutshall, D. Maley,
`A. DelVecchio, R. Macarron, G. A. Hofmann, Z. Alnoah, H. Y. Cheng, G.
`Chan, S. Khandekar, R. M. Keenan, and R. T. Sarisky. 2002. Identification
`and biological characterization of heterocyclic inhibitors of the hepatitis C
`virus RNA-dependent RNA polymerase. J. Biol. Chem. 277:38322–38327.
`10. Eldrup, A. B., M. Prhavc, J. Brooks, B. Bhat, T. P. Prakash, Q. Song, S. Bera,
`N. Bhat, P. Dande, P. D. Cook, C. F. Bennett, S. S. Carroll, R. G. Ball, M.
`Bosserman, C. Burlein, L. F. Colwell, J. F. Fay, O. A. Flores, K. Getty, R. L.
`Lafemina, J. Leone, M. MacCoss, D. R. McMasters, J. E. Tomassini, L. D.
`Von, B. Wolanski, and D. B. Olsen. 2004. Structure-activity relationship of
`heterobase-modified 2⬘-C-methyl ribonucleosides as inhibitors of hepatitis C
`virus RNA replication. J. Med. Chem. 47:5284–5297.
`11. Fried, M. W., M. L. Shiffman, K. R. Reddy, C. Smith, G. Marinos, F. L.
`Goncales, Jr., D. Haussinger, M. Diago, G. Carosi, D. Dhumeaux, A. Craxi,
`A. Lin, J. Hoffman, and J. Yu. 2002. Peginterferon alfa-2a plus ribavirin for
`chronic hepatitis C virus infection. N. Engl. J. Med. 347:975–982.
`12. Gopalsamy, A., A. Aplasca, G. Ciszewski, K. Park, J. W. Ellingboe, M.
`Orlowski, B. Feld, and A. Y. Howe. 2006. Design and synthesis of 3,4-dihydro-
`1H-[1]-benzothieno[2,3-c]pyran and 3,4-dihydro-1H-pyrano[3,4-b]benzofuran
`derivatives as non-nucleoside inhibitors of HCV NS5B RNA dependent RNA
`polymerase. Bioorg. Med. Chem. Lett. 16:457–460.
`13. Gopalsamy, A., K. Lim, G. Ciszewski, K. Park, J. W. Ellingboe, J. Bloom, S.
`Insaf, J. Upeslacis, T. S. Mansour, G. Krishnamurthy, M. Damarla, Y.
`Pyatski, D. Ho, A. Y. Howe, M. Orlowski, B. Feld, and J. O’Connell. 2004.
`Discovery of pyrano[3,4-b]indoles as potent and selective HCV NS5B poly-
`merase inhibitors. J. Med. Chem. 47:6603–6608.
`14. Guo, J. T., V. V. Bichko, and C. Seeger. 2001. Effect of alpha interferon on
`the hepatitis C virus replicon. J. Virol. 75:8516–8523.
`15. Hinrichsen, H., Y. Benhamou, H. Wedemeyer, M. Reiser, R. E. Sentjens,
`J. L. Calleja, X. Forns, A. Erhardt, J. Cronlein, R. L. Chaves, C. L. Yong, G.
`Nehmiz, and G. G. Steinmann. 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.
`16. Lamarre, D., P. C. Anderson, M. Bailey, P. Beaulieu, G. Bolger, P. Bonneau,
`M. Bos, D. R. Cameron, M. Cartier, M. G. Cordingley, A. M. Faucher, N.
`Goudreau, S. H. Kawai, G. Kukolj, L. Lagace, S. R. LaPlante, H. Narjes,
`M. A. Poupart, J. Rancourt, R. E. Sentjens, G. R. St, B. Simoneau, G.
`Steinmann, D. Thibeault, Y. S. Tsantrizos, S. M. Weldon, C. L. Yong, and M.
`Llinas-Brunet. 2003. An NS3 protease inhibitor with antiviral effects in
`humans infected with hepatitis C virus. Nature 426:186–189.
`17. Lin, C., C. A. Gates, B. G. Rao, D. L. Brennan, J. R. Fulghum, Y. P. Luong,
`J. D. Frantz, K. Lin, S. Ma, Y. Y. Wei, R. B. Perni, and A. D. Kwong. 2005.
`In vitro studies of cross-resistance mutations against two hepatitis C virus
`serine protease inhibitors, VX-950 and BILN 2061. J. Biol. Chem. 280:
`36784–36791.
`18. Lin, C., K. Lin, Y. P. Luong, B. G. Rao, Y. Y. Wei, D. L. Brennan, J. R.
`Fulghum, H. M. Hsiao, S. Ma, J. P. Maxwell, K. M. Cottrell, R. B. Perni,
`C. A. Gates, and A. D. Kwong. 2004. In vitro resistance studies of hepatitis C
`virus serine protease inhibitors, VX-950 and BILN 2061: structural analysis
`indicates different resistance mechanisms. J. Biol. Chem. 279:17508–17514.
`19. Lin, K., A. D. Kwong, and C. Lin. 2004. Combination of a hepatitis C virus
`NS3-NS4A protease inhibitor and alpha interferon synergistically inhibits
`viral RNA replication and facilitates viral RNA clearance in replicon cells.
`Antimicrob. Agents Chemother. 48:4784–4792.
`20. Lin, K., R. B. Perni, A. D. Kwong, and C. Lin. 2006. VX-950, a novel hepatitis
`C virus (HCV) NS3-4A protease inhibitor, exhibits potent antiviral activities
`in HCv replicon cells. Antimicrob. Agents Chemother. 50:1813–1822.
`21. Lu, L., T. J. Pilot-Matias, K. D. Stewart, J. T. Randolph, R. Pithawalla, W.
`He, P. P. Huang, L. L. Klein, H. Mo, and A. Molla. 2004. Mutations con-
`ferring resistance to a potent hepatitis C virus serine protease inhibitor in
`vitro. Antimicrob. Agents Chemother. 48:2260–2266.
`22. Ludmerer, S. W., D. J. Graham, E. Boots, E. M. Murray, A. Simcoe, E. J.
`
`Markel, J. A. Grobler, O. A. Flores, D. B. Olsen, D. J. Hazuda, and R. L.
`LaFemina. 2005. Replication fitness and NS5B drug sensitivity of diverse
`hepatitis C virus isolates characterized by using a transient replication assay.
`Antimicrob. Agents Chemother. 49:2059–2069.
`23. Manns, M. P., J. G. McHutchison, S. C. Gordon, V. K. Rustgi, M. Shiffman,
`R. Reindollar, Z. D. Goodman, K. Koury, M. Ling, and J. K. Albrecht. 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.
`24. Migliaccio, G., J. E. Tomassini, S. S. Carroll, L. Tomei, S. Altamura, B.
`Bhat, L. Bartholomew, M. R. Bosserman, A. Ceccacci, L. F. Colwell, R.
`Cortese, F. R. De, A. B. Eldrup, K. L. Getty, X. S. Hou, R. L. LaFemina, S. W.
`Ludmerer, M. MacCoss, D. R. McMasters, M. W. Stahlhut, D. B. Olsen,
`D. J. Hazuda, and O. A. Flores. 2003. Characterization of resistance to
`non-obligate chain-terminating ribonucleoside analogs that inhibit hepatitis
`C virus replication in vitro. J. Biol. Chem. 278:49164–49170.
`25. Neumann, A. U., N. P. Lam, H. Dahari, D. R. Gretch, T. E. Wiley, T. J.
`Layden, and A. S. Perelson. 1998. Hepatitis C viral dynamics in vivo and the
`antiviral efficacy of interferon-alpha therapy. Science 282:103–107.
`26. Nguyen, T. T., A. T. Gates, L. L. Gutshall, V. K. Johnston, B. Gu, K. J. Duffy,
`and R. T. Sarisky. 2003. Resistance profile of a hepatitis C virus RNA-
`dependent RNA polymerase benzothiadiazine inhibitor. Antimicrob. Agents
`Chemother. 47:3525–3530.
`27. Olsen, D. B., A. B. Eldrup, L. Bartholomew, B. Bhat, M. R. Bosserman, A.
`Ceccacci, L. F. Colwell, J. F. Fay, O. A. Flores, K. L. Getty, J. A. Grobler,
`R. L. LaFemina, E. J. Markel, G. Migliaccio, M. Prhavc, M. W. Stahlhut,
`J. E. Tomassini, M. MacCoss, D. J. Hazuda, and S. S. Carroll. 2004. A
`7-deaza-adenosine analog is a potent and selective inhibitor of hepatitis C
`virus replication with excellent pharmacokinetic properties. Antimicrob.
`Agents Chemother. 48:3944–3953.
`28. Perelson, A. S., A. U. Neumann, M. Markowitz, J. M. Leonard, and D. D. Ho.
`1996. HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span,
`and viral generation time. Science 271:1582–1586.
`29. Reesink, H., S. Zeuzem, C. Weegink, N. Forestier, A. van Vilet, J. van de
`Wetering de Rooij, L. McNair, S. Purdy, J. Chu, and P. Jansen. 2005. Final
`results of a phase 1B, multiple-dose study of VX-950, a hepatitis C virus
`protease inhibitor. Hepatology 42:234A–235A.
`30. Roberts, S., G. Cooksley, G. Dore, R. Robson, D. Shaw, H. Berns, M. Brandl,
`S. Fettner, G. Hill, E. Ipe, K. Klumpp, M. Mannino, E. O’Mara, I. Najera,
`Y. Tu, and C. Washingtion. 2006. Results of a phase 1B, multiple dose study
`of R1626, a novel nucleoside analog targeting HCV polymerase in chronic
`HCV genotype 1 patients. Hepatology 44:692A.
`31. Sarrazin, C., T. Kieffer, D. Bartels, B. Hanzelka, U. Muh, M. Welker, D.
`Wincheringer, C. Lin, T. Grossman, S. Purdy, C. Weegink, H. Reesink, S.
`Zeuzem, and A. D. Kwong. 2005. Characterization of viral variants in the
`HCV NS3 protease domain of genotype 1 patients that are selected during
`14 days of dosing with VX-950. Hepatology 42:751A.
`32. Shimakami, T., M. Hijikata, H. Luo, Y. Y. Ma, S. Kaneko, K. Shimotohno,
`and S. Murakami. 2004. Effect of interaction between hepatitis C virus
`NS5A and NS5B on hepatitis C virus RNA replication with the hepatitis C
`virus replicon. J. Virol. 78:2738–2748.
`33. Tanabe, Y., N. Sakamoto, N. Enomoto, M. Kurosaki, E. Ueda, S. Maekawa,
`T. Yamashiro, M. Nakagawa, C. H. Chen, N. Kanazawa, S. Kakinuma, and
`M. Watanabe. 2004. Synergistic inhibition of intracellular hepatitis C virus
`replication by combination of ribavirin and interferon-alpha. J. Infect. Dis.
`189:1129–1139.
`34. Trozzi, C., L. Bartholomew, A. Ceccacci, G. Biasiol, L. Pacini, S. Altamura,
`F. Narjes, E. Muraglia, G. Paonessa, U. Koch, F. R. De, C. Steinkuhler, and
`G. Migliaccio. 2003. In vitro selection and characterization of hepatitis C
`virus serine protease variants resistant to an active-site peptide inhibitor.
`J. Virol. 77:3669–3679.
`35. Yi, M., X. Tong, A. Skelton, R. Chase, T. Chen, A. Prongay, S. L. Bogen, A. K.
`Saksena, F. G. Njoroge, R. L. Veselenak, R. B. Pyles, N. Bourne, B. A.
`Malcolm, and S. M. Lemon. 2006. Mutations conferring resistance to SCH6,
`a novel hepatitis C virus NS3/4A protease inhibitor. Reduced RNA replica-
`tion fitness and partial rescue by second-site mutations. J. Biol. Chem.
`281:8205–8215.
`36. Zeuzem, S., C. Sarrazin, R. Rouzier, A. Tarral, N. Brion, N. Forestier, S.
`Gupta, D. Deckman, K. Fellows, M. Hussain, D. Cutler, and J. Zhang. 2005.
`Anti-viral activity of SCH 503034, a HCV protease inhibitor, administered as
`monotherapy in hepatitis C genotype-1 patients refractory to pegylated in-
`terferon. Hepatology 42:233A–234A.
`
`

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