01.03.2014 Views

GLoBAL ANTIVIRAL JoURNAL - IHL Press

GLoBAL ANTIVIRAL JoURNAL - IHL Press

GLoBAL ANTIVIRAL JoURNAL - IHL Press

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

GAJ<br />

Gl o b a l<br />

An t i v i r a l<br />

Jo u r n a l<br />

December 9-13, 2007<br />

The Westin Maui<br />

Lahaina, Hawaii<br />

Final Program and Abstract Book<br />

This program is sponsored by Emory University School of Medicine<br />

GAJ<br />

Volume 3, Supplement 2


GAJ<br />

Gl o b a l<br />

An t i v i r a l<br />

Jo u r n a l<br />

Aims and Scope<br />

Global Antiviral Journal publishes peer-reviewed original works related to international efforts to advance antiviral<br />

discovery and development, including full-length articles and short papers, as well as solicited review articles, conference<br />

reports, letters and book reviews. Occasional supplements contain conference abstracts presentations and/or posters<br />

from international meetings in the fields of virology and antiviral research. The scope of the journal encompasses<br />

chemistry and biological advances in the fundamental and clinical study of antiviral diseases and their treatment. Areas<br />

covered include HIV, hepatitis B, hepatitis C and emerging viruses, co-infections, vaccines, animal models, pharmacology,<br />

microbicides, alternative therapies, viral dynamics and resistance issues.<br />

The journal is published online by <strong>IHL</strong> <strong>Press</strong> at www.ihlpress.com/gaj.html.<br />

All printed supplements are also made available online.<br />

Publication Policy<br />

Global Antiviral Journal publishes only original, documented research of high scientific quality, following accepted ethical<br />

standards of research. Submission of a manuscript signifies that it has been neither copyrighted, published, nor submitted<br />

or accepted for publication elsewhere.<br />

Editor-in-Chief<br />

Raymond F. Schinazi, Emory University School of Medicine and Veterans Affairs Medical Center, Department of Pediatrics,<br />

Medical Research 151H, 1670 Clairmont Road, Decatur, Georgia, 30033, USA<br />

Editorial Office<br />

<strong>IHL</strong> <strong>Press</strong>, 26 Bailey Road, Arlington, MA, 02476, USA<br />

Telephone: +1 781 648 1933<br />

Fax: +1 781 646 2699<br />

info@ihlpress.com<br />

Subscription Details<br />

Subscription prices are available upon request from the Publisher. All inquiries should be directed to the Editorial Office.<br />

Advertising and Supplements<br />

All advertising enquiries and supplement proposals, including advertising within supplements, should be directed to the<br />

Editorial Office.<br />

Copyright<br />

© 2007 <strong>IHL</strong> <strong>Press</strong>. All rights reserved.<br />

No part of this work may be reproduced, stored in a retrieval system or transmitted in any form or by any means,<br />

electronic, mechanical, photocopying, recording or otherwise, without prior written permission of the Publisher.<br />

Notice<br />

No responsibility is assumed by the Publisher for any injury and/or damage to persons or property as a matter of products<br />

liability, negligence or otherwise, or from any use or operation of any methods, products, diagnoses, drug dosages,<br />

instructions or ideas contained in the material herein.<br />

ISSN (print): 1556-9047<br />

ISSN (online): 1556-9055


December 9-13, 2007 • The Westin Maui • Lahaina, Hawaii<br />

Final Program<br />

and Abstract Book<br />

This program is sponsored by Emory University School of Medicine<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

i


Table of Contents<br />

Corporate Supporters .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . iv<br />

Continuing Medical Education............................................................... v<br />

Scholarships .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v<br />

Conference Committees .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vi<br />

Special Events........................................................................... vii<br />

Scientific Program .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix<br />

Abstracts.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xvii<br />

Page<br />

Sunday, December 9<br />

State of the Art Lectures.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1<br />

Monday, December 10<br />

Advances in New Models for HBV and HCV..........................................5<br />

Tuesday, December 11<br />

Diagnosis and New Treatments for HBV and HCV....................................19<br />

Hepatitis B Foundation Symposium:<br />

Early Detection and Management of Cirrhosis and Hepatocellular Carcinoma..........39<br />

Immunology & Pathogenesis.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47<br />

Wednesday, December 12<br />

New Therapeutic Approaches.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57<br />

Resistance to Antiviral Agents.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81<br />

Co-infection with HIV and Other Viruses .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95<br />

Thursday, December 13<br />

Pharmacology and Drug Interactions of HBV and HCV Therapeutics.. . . . . . . . . . . . . . . . . 105<br />

Optimizing the Outcome of Therapeutics for HBV and HCV .. . . . . . . . . . . . . . . . . . . . . . . . 113<br />

Late Breaker Session .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127<br />

Author Index........................................................................... 133<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

iii


Support for HEP DART 2007 was provided by:<br />

Platinum Support<br />

Gold Support<br />

Silver Support<br />

Bronze Support<br />

Anadys Pharmaceuticals Inc. • GlaxoSmithKline • GlaxoSmithKline R&D<br />

Merchant & Gould LLC • Romark Laboratories, L.C. • Samchully Pharmaceutical Co., Ltd.<br />

Additional Support<br />

ACLIRES International Ltd • Department of Veterans Affairs<br />

Genelabs Technologies, Inc. • GlobeImmune, Inc. • Novartis Vaccines and Diagnostics, Inc.<br />

Raymond F. Schinazi & Family Foundation • RFS Pharma, LLC<br />

William H. Prusoff Foundation<br />

iv Global Antiviral Journal Volume 3, Supplement 2


Continuing Medical Education<br />

HEP DART 2007 is sponsored by Emory University School of Medicine.<br />

Accreditation Statement<br />

The Emory University School of Medicine is accredited by the ACCME to provide continuing medical<br />

education for physicians.<br />

Continuing Medical Education<br />

The Emory University School of Medicine designates this educational activity for a maximum of<br />

23.5 AMA PRA Category 1 Credits. Physicians should only claim credit commensurate with the<br />

extent of their participation in the activity.<br />

Conference Objectives<br />

The Scientific Committee has designed the Conference program to ensure that the delegates achieve<br />

the following objectives:<br />

• Understand the role of viral targets in the drug development and discovery process<br />

• Identify the next generation of inhibitors of viral hepatitis<br />

• Assess the impact of resistance and treatment failure in the drug development and<br />

discovery process<br />

• Increase awareness of the clinical impact of antiviral agents<br />

• Understand the consequences of co-infection with HIV on the management of patients<br />

• Assess the role of vaccines and therapeutic vaccines in future therapies for viral hepatitis<br />

Scholarships<br />

Conference scholarships were provided for post-doctoral fellows, nurses, assistant professors,<br />

underrepresented minorities, and residents of developing countries.<br />

2007 Recipients<br />

Cafer Eroglu, Ondokuz Mayis University, Turkey<br />

Pablo Gastaminza, The Scripps Research Institute, USA<br />

Jason Grebely, National University of New South Wales, Australia<br />

Gustine Liu-Young, Yale University School of Medicine, USA<br />

Severine Margeridon-Thermet, Stanford University, USA<br />

Saneyuki Ujino, Chiba Institute of Technology, Japan<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

v


Organizing Committee<br />

Eugene Schiff, Chair<br />

University of Miami, USA<br />

Raymond F. Schinazi, Chair<br />

Emory University/Veteran Affairs Medical Center, USA<br />

Robert Murphy, Co-chair<br />

Northwestern University, USA<br />

Charles Rice, Co-chair<br />

The Rockefeller University, USA<br />

Emeritus Chair and Founding Member<br />

Jean-Pierre Sommadossi<br />

Idenix Pharmaceuticals, USA<br />

Miriam Alter<br />

University of Texas Medical Branch at Galveston, USA<br />

Scientific Advisory Committee<br />

Stanley M. Lemon<br />

University of Texas Medical Branch at Galveston, USA<br />

Mithat Bozdayi<br />

Ankara University, Turkey<br />

Ralf Bartenschlager<br />

University of Heidelberg, Germany<br />

Frank V. Chisari<br />

The Scripps Research Institute, USA<br />

Jules Dienstag<br />

Harvard School of Medicine, USA<br />

Michael W. Fried<br />

University of North Carolina at Chapel Hill, USA<br />

Robert Gish<br />

California Pacific Medical Center, USA<br />

Allison Jilbert<br />

University of Adelaide, Australia<br />

Stephen Locarnini<br />

Victorian Infectious Diseases Reference Laboratory, Australia<br />

Anna Lok<br />

University of Michigan Medical Center, USA<br />

Michael P. Manns<br />

Hannover Medical School, Germany<br />

John G. McHutchison<br />

Duke University, USA<br />

Masao Omata<br />

University of Tokyo, Japan<br />

Thierry Poynard<br />

Groupe Hospitalier Pitié-Salpêtrière, France<br />

Lorne Tyrrell<br />

University of Alberta, Canada<br />

Brent E. Korba<br />

Georgetown University, USA<br />

vi Global Antiviral Journal Volume 3, Supplement 2


Special Events<br />

Satellite Symposium<br />

Toward Curative Therapies for Hepatitis C<br />

Sunday, December 9, 2007<br />

9:00<br />

<br />

Welcome Reception<br />

Sunday, December 9, 2007<br />

18:00<br />

Lanai Pool Deck, Westin Maui<br />

<br />

Poster Session Reception<br />

Tuesday, December 11, 2007<br />

16:30<br />

Haleakala Ballroom<br />

<br />

Gala Party<br />

Wednesday, December 12, 2007<br />

19:30<br />

Aloha Pavilion, Westin Maui<br />

Conference Secretariat<br />

1631 Phoenix Boulevard, Suite 4<br />

College Park, Georgia 30349 USA<br />

Telephone: +1 770 997 2484<br />

Facsimile: +1 770 997 2488<br />

E-mail: info@informedhorizons.com<br />

Website: www.informedhorizons.com<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

vii


HEP DART 2007<br />

Frontiers in Drug Development for Viral Hepatitis<br />

Scientific Program<br />

Sunday, December 9, 2007<br />

9:00 Satellite Symposium: Toward Curative Therapies for Hepatitis C<br />

Abstract<br />

12:10 Satellite Symposium Lunch<br />

HEP DART 2007 Opening Session<br />

15:00 Opening Remarks<br />

Raymond F. Schinazi<br />

Emory University/Veteran Affairs Medical Center, USA<br />

Chairs: Robert Murphy<br />

Charles Rice<br />

Northwestern University, USA<br />

The Rockefeller University, USA<br />

15:15 State of the Art Lecture<br />

The Changing Global Epidemiology of HBV and HCV 01<br />

Kevin Fenton<br />

Centers for Disease Control and Prevention, USA<br />

16:00 State of the Art Lecture<br />

New Insights into Hepatitis B Pathogenesis and Resistance 02<br />

Stephen Locarnini<br />

Victorian Infectious Diseases Reference Laboratory, Australia<br />

18:00 Welcome Reception<br />

Monday, December 10, 2007<br />

Chairs: Raymond F. Schinazi<br />

Robert Murphy<br />

Emory University/Veteran Affairs Medical Center, USA<br />

Northwestern University, USA<br />

8:00 Presentation of William H. Prusoff HEP DART Lifetime Achievement Award<br />

Raymond F. Schinazi<br />

Emory University/Veteran Affairs Medical Center, USA<br />

Robert Murphy<br />

Northwestern University, USA<br />

8:10 William H. Prusoff HEP DART Lifetime Achievement Award<br />

A Pre-Mortem Lookback Investigation of One’s Life in Research: 03<br />

The Payoffs of a Persistent Patient<br />

Harvey Alter<br />

National Institutes of Health, USA<br />

8:50 State of the Art Lecture<br />

New Insights into the HCV Replication Cycle: Lessons Learned from Novel Culture Systems 04<br />

Ralf Bartenschlager<br />

University of Heidelberg, Germany<br />

Advances in New Models for HBV and HCV<br />

9:20 HCV Cell Culture Systems and the Development of Antiviral Therapeutics 05<br />

T. Jake Liang National Institutes of Health, USA<br />

9:40 Role of Negative Regulatory Signals in the Pathogenesis of HCV 06<br />

Arash Grakoui<br />

Emory Vaccine Center, USA<br />

10:00 A Model System for Hepatitis B and C Virus Co-infection 07<br />

Volker Brass<br />

University of Freiburg, Germany<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

ix


Abstract<br />

10:20 Special Lecture<br />

Insights into the Impact of HBV and HCV Viral Dynamics on Antiviral Therapy 08<br />

Alan S. Perelson<br />

Los Alamos National Laboratories, USA<br />

10:40 Panel Discussion<br />

11:05 Break<br />

Oral Abstract Presentations<br />

Chairs: Thierry Poynard<br />

Brent E. Korba<br />

Groupe Hospitalier Pitié-Salpêtrière, France<br />

Georgetown University, USA<br />

11:30 Interferon Modulation of Cellular microRNAs as a Novel Antiviral Mechanism 09<br />

Irene Pedersen<br />

University of California, San Diego, USA<br />

11:40 DNA Microarray Analysis of the NS3 and NS5B Genes from Hepatitis C Genotype 1a 10<br />

Infected Patients<br />

Gustine Liu-Young<br />

Yale University, USA<br />

11:50 Assessment of Both Virological Response at Week 4 and at Week 12 Optimizes Prediciton of 11<br />

Treatment Outcome in Patients with Chronic Hepatitis C Treated with Peginterferon Alfa-2B<br />

plus Ribavirin<br />

Michelle Martinot-Peignoux INSERM U-773-CRB3, France<br />

12:00 Week 24 is the Optimal Time Point for Predicting Outcomes at 2 Years with Telbivudine 12<br />

Rajender Reddy<br />

University of Pennsylvania School of Medicine, USA<br />

12:10 Lunch<br />

Monday Evening Session<br />

The Business of Hepatitis Antivirals<br />

Chairs: Abel De La Rosa<br />

Patrick Higgins<br />

Pharmasset, Inc., USA<br />

Pharmasset, Inc., USA<br />

17:00 The Business of Hepatitis Antivirals: Bridging Diverse Interests<br />

Patrick Higgins<br />

Pharmasset, Inc., USA<br />

17:20 Panel Discussion<br />

Adam Cutler<br />

Sandra Lehrman<br />

Dennis Liotta<br />

Richard Smith<br />

Frank Zavrl<br />

Canaccord Adams, USA<br />

Merck Research Laboratories, USA<br />

Emory University, USA<br />

JP Morgan Securities, Inc., USA<br />

Adage Capital, USA<br />

Tuesday, December 11, 2007<br />

Diagnosis and New Treatments for HBV and HCV<br />

Chairs: Ralf Bartenschlager<br />

University of Heidelberg, Germany<br />

Charles Rice<br />

The Rockefeller University, USA<br />

8:00 State of the Art Lecture<br />

HCV Replicons vs. Infectious Virus Systems in Drug Discovery 18<br />

Stanley M. Lemon<br />

University of Texas Medical Branch at Galveston, USA<br />

8:30 State of the Art Lecture<br />

The Size of the Viral Inoculum Determines the Kinetics, Magnitude and Outcome 19<br />

of Hepatitis B Virus Infection<br />

Frank V. Chisari<br />

Scripps Research Institute, USA<br />

x Global Antiviral Journal Volume 3, Supplement 2


Abstract<br />

9:00 Population Genetics of HBV and HCV Quasispecies: Implications for Drug Development 20<br />

and Drug-resistance Testing<br />

Robert W. Shafer<br />

Stanford University, USA<br />

9:20 Nitazoxanide: A Potent Antiviral Agent against HCV and HBV 21<br />

Brent E. Korba<br />

Georgetown University, USA<br />

9:40 Clinical Implications of Combined Intra-cellular and Cellular Evolution of HCV Resistance 22<br />

during Direct Anti-HCV Treatment<br />

Avidan U. Neumann<br />

Bar-Ilan University, Israel<br />

10:00 Panel Discussion<br />

10:30 Break<br />

Oral Abstract Presentations<br />

Chairs: George Lau<br />

Adrian DiBisceglie<br />

Queen Mary Hospital, Hong Kong<br />

Saint Louis University School of Medicine, USA<br />

11:00 Phase 2 Studies with Albinterferon Alfa-2b (alb-IFN) Dosed q4wk Provide Insights 23<br />

into Dose Selection for Future Studies<br />

Ira Jacobson<br />

Weill Medical College of Cornell University, USA<br />

11:10 Robust Antiviral Efficacy of a “Finger-loop” Allosteric Inhibitor of the HCV Polymerase 24<br />

in HCV Infected Chimpanzees<br />

Raffaele De Francesco<br />

I.R.B.M. “P Angeletti,” Italy<br />

11:20 Preclinical Development of the Amphipathic DNA Polymer REP 9AC for the Treatment 25<br />

of HBV Infection<br />

Andrew Vaillant<br />

REPLICor Inc., Canada<br />

11:30 Inhibition of Hepatitis C Virus Replication by Octadecyloxypropyl-(S)-HPMPA 26<br />

in Genotype 1B and 2A Replicons<br />

Karl Y. Hostetler<br />

University of California, San Diego, USA<br />

11:40 FKBP8 Plays a Crucial Role in the Replication of Hepatitis C Virus 27<br />

Yoshiharu Matsuura<br />

Osaka University, Japan<br />

11:50 Initial Recommendations for HCV Drug Resistance Analysis: A Consensus Statement 28<br />

from the HCV Drug Resistance Advisory Group<br />

Kai Lin<br />

Novartis Institutes for Biomedical Research, Inc., USA<br />

12:00 Lunch<br />

Hepatitis B Foundation Symposium: Early Detection and Management of Cirrhosis<br />

and Hepatocellular Carcinoma<br />

Chairs: Timothy Block<br />

Hepatitis B Foundation and Drexel University<br />

Medical College, USA<br />

Harvey Alter<br />

National Institutes of Health, USA<br />

13:30 Introduction: Early Detection and Management of Cirrhosis and Hepatocellular Carcinoma<br />

Timothy Block<br />

Hepatitis B Foundation and Drexel University<br />

Medical College, USA<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xi


Abstract<br />

13:40 Hepatocellular Carcinoma: Why Early Diagnosis is Needed 39<br />

Adrian M. DiBisceglie<br />

Saint Louis University Liver Center, USA<br />

13:55 Assessment of Fibrosis: Non Biopsy Methods to Determine Hepatic Fibrosis 40<br />

Nezam H. Afdhal<br />

Harvard Medical School, USA<br />

14:10 Screening Fibrosis: “La Révolution Française” 41<br />

Thierry Poynard<br />

Groupe Hospitalier Pitié-Salpêtrière, France<br />

14:25 Treatment of HCC over the Past Decade: The Experience of over Four Thousand Patients 42<br />

in Japan<br />

Masao Omata<br />

University of Tokyo, Japan<br />

14:40 Use of Pre-emptive Nucleoside Analogue Therapy for Hepatitis B Reactivation 43<br />

after Chemotherapy<br />

George K. K. Lau<br />

Queen Mary Hospital, Hong Kong<br />

14:55 Panel Discussion<br />

16:30 Poster Session<br />

Tuesday Evening Session<br />

Immunology & Pathogenesis<br />

Chairs Lawrence M. Blatt<br />

Michael Fried<br />

Alios BioPharma, Inc., USA<br />

University of North Carolina at Chapel Hill, USA<br />

18:00 State of the Art Lecture<br />

Cellular Immune Responses against Hepatitis C in Acute and Chronic Infection 44<br />

Margaret J. Koziel<br />

Harvard Medical School, USA<br />

18:30 Broadly Neutralizing Antibodies to HCV: Definition of Epitopes and Antiviral Activity 45<br />

In Vitro and In Vivo<br />

Mansun Law<br />

The Scripps Research Institute, USA<br />

18:50 Innate Immune Studies in Hepatitis B: Novel Studies in Pathogenesis and Treatment 46<br />

Kumar Visvanathan<br />

Monash Institute of Medical Research, Australia<br />

19:10 Early Steps in the Replicative Cycle of Hepatitis B Virus 47<br />

Dieter Glebe<br />

University of Giessen, Germany<br />

19:30 Panel Discussion<br />

Wednesday, December 12, 2007<br />

New Therapeutic Approaches I<br />

Chairs: Stanley M. Lemon<br />

Karen Anderson<br />

University of Texas Medical Branch at Galveston, USA<br />

Yale University School of Medicine, USA<br />

8:00 State of the Art Lecture<br />

HCV Entry Pathways and Implications for the Development of Entry Inhibitors 53<br />

Matthew J. Evans<br />

The Rockefeller University, USA<br />

8:30 The Role of CD81 and Scavenger Receptor Class B Member 1 in HCV Entry 54<br />

Peter Balfe<br />

University of Birmingham, UK<br />

8:50 Lipid Droplet is an Important Organelle for Production of Infectious Hepatitis C Virus 55<br />

Kunitada Shimotohno<br />

Keio University, Japan<br />

xii Global Antiviral Journal Volume 3, Supplement 2


Abstract<br />

9:10 The Role of Cyclophilins and Cyclophilin Inhibitors in the Replication of HCV 56<br />

Rafael Crabbé<br />

Debiopharm S.A., Switzerland<br />

Oral Abstract Presentation<br />

9:30 HCV Infection Increases Claudin-1 and Claudin-7 Expression by Inducing Cirrhosis 57<br />

Andras Kiss<br />

Semmelweis Medical University, Hungary<br />

9:40 Panel Discussion<br />

10:05 Break<br />

New Therapeutic Approaches II<br />

Chairs: Yves Benhamou<br />

Marion Peters<br />

Hôpital Pitié-Salpêtrière, France<br />

University of California, San Francisco, USA<br />

Oral Abstract Presentations<br />

10:30 Development of Novel Hyperglycosylated Type 1 Interferons: A Strategy to Improve PK 58<br />

Performance without Loss of Biological Potency<br />

Lawrence M. Blatt<br />

Alios BioPharma, Inc., USA<br />

10:40 Bioinformatics Resources Supporting the Analysis of Hepatitis C Virus 59<br />

Elliot J. Lefkowitz<br />

University of Alabama at Birmingham, USA<br />

Invited Presentations<br />

10:50 A Combination of Direct Antiviral Compounds can Achieve Sustained Viral Response 60<br />

in Hepatitis C Virus-infected Chimpanzees<br />

David B. Olsen<br />

Merck Research Laboratories, USA<br />

11:10 Potent Antiviral Activity of the Nucleoside HCV Inhibitor, R7128, in Prior 61<br />

IFN Non-responders<br />

John G. McHutchison<br />

Duke University, USA<br />

11:30 R1626 Demonstrates Synergistic Antiviral Effect in Combination with Peginterferon Alfa-2a 62<br />

[40 KD], with or without Ribavirin, and High Barrier to Resistance Development<br />

David Nelson<br />

University of Florida, USA<br />

11:50 Tenofovir DF (TDF) Showed Superior Antiviral Efficacy to Adefovir Dipivoxil (ADV) 63<br />

at 48 Weeks in Two Pivotal, Randomized, Double-Blind, Studies for the Treatment of<br />

HBeAg-Negative (-) and HBeAg-Positive (+) Chronic Hepatitis B (CHB):<br />

Study 102 and Study 103<br />

Patrick Marcellin<br />

Hôpital Beaujon, France<br />

12:10 Panel Discussion<br />

12:35 Lunch<br />

wednesday Evening Session<br />

Resistance to Antiviral Agents<br />

Chairs: Patrick Marcellin<br />

Michael P. Manns<br />

Hôpital Beaujon, France<br />

Hannover Medical School, Germany<br />

16:00 State of the Art Lecture<br />

Navigating Dangerous Complexities: Antiviral Strategies for Clinical Trials for Hepatitis C<br />

Yves Benhamou<br />

Hôpital Pitié-Salpêtrière, France<br />

16:30 Overcoming HCV Treatment-resistant Characteristics 77<br />

Michael W. Fried<br />

University of North Carolina at Chapel Hill, USA<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xiii


Abstract<br />

16:50 Genetic and Structural Variability of the Hepatitis C Viral Polymerase, NS5B: Implications 78<br />

for Resistance to Inhibitors<br />

Philip C. Simister<br />

CNRS, France<br />

17:10 Panel Discussion<br />

Co-infection with HIV and Other Viruses<br />

Chairs: Robert Gish<br />

California Pacific Medical Center, USA<br />

Masao Omata<br />

University of Tokyo, Japan<br />

17:30 Treatment of HBV/HIV Co-infections 88<br />

Marion Peters<br />

University of California, San Francisco, USA<br />

17:50 Treatment of HCV in HIV-infected Individuals 89<br />

Zelalem Temesgen<br />

Mayo Clinic, USA<br />

18:10 HCV/HIV Co-infections: Challenges from the Perspective of TAG 90<br />

Tracy Swan<br />

Treatment Action Group (TAG), USA<br />

18:30 Panel Discussion<br />

20:00 Gala Party<br />

Thursday, December 13, 2007<br />

Pharmacology and Drug Interactions of HBV and HCV Therapeutics<br />

Chairs: Dennis Liotta<br />

Emory University, USA<br />

Nezam H. Afdhal<br />

Harvard Medical School, USA<br />

8:00 State of the Art Lecture<br />

STAT-C: Role in HCV Treatment Algorithm in 2008<br />

John G. McHutchison<br />

Duke University, USA<br />

8:30 Preclinical Drug Metabolism in Viral Hepatitis Drug Discovery 97<br />

Adrian S. Ray<br />

Gilead Sciences, Inc., USA<br />

8:50 Frontiers of Clinical Pharmacology in Hepatitis Drug Development 98<br />

Charles W. Flexner<br />

Johns Hopkins University, USA<br />

9:10 Special Lecture<br />

Current and Future: HCV Maintenance Therapy 99<br />

Karen L. Lindsay<br />

University of Southern California, USA<br />

9:30 Break<br />

Late Breaker Abstracts<br />

Chairs: Margaret J. Koziel<br />

Stephen Locarnini<br />

Harvard Medical School, USA<br />

Victorian Infectious Diseases Reference Laboratory, Australia<br />

10:00 State of the Art Lecture - Part I<br />

Long Term Follow-up of HCV Patients Completing Peg-IFN Plus Ribavirin Treatment - 115<br />

Is There a Cure?<br />

Michael P. Manns<br />

Hannover Medical School, Germany<br />

xiv Global Antiviral Journal Volume 3, Supplement 2


Abstract<br />

10:20 State of the Art Lecture - Part II<br />

Delta Hepatitis: Treatment Options for a Largely Neglected Disease 116<br />

Michael P. Manns<br />

Hannover Medical School, Germany<br />

10:40 Biochemical Mechanism of Entecavir as an Antiviral Polymerase Inhibitor 117<br />

Karen S. Anderson<br />

Yale University School of Medicine, USA<br />

11:00 Advancing RNAi-based Therapeutic from Discovery to Clinic 118<br />

Robert Gish<br />

California Pacific Medical Research Institute, USA<br />

Catherine Pachuk<br />

Nucleonics Inc., USA<br />

11:20 Panel Discussion<br />

11:50 Reframing the Knowledge and Looking Beyond the Horizon<br />

Eugene Schiff<br />

University of Miami, USA<br />

12:10 Closing Remarks<br />

Raymond F. Schinazi<br />

Emory University/Veteran Affairs Medical Center, USA<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xv


Abstracts<br />

page session title and author abstract<br />

1 State of the Art Lectures<br />

3 The Changing Global Epidemiology of HBV and HCV 01<br />

K Fenton<br />

3 New Insights into Hepatitis B Pathogenesis and Resistance 02<br />

S Locarnini<br />

5 Advances in New Models for HBV and HCV<br />

7 A Pre-mortem Lookback Investigation of One's Life in Research: 03<br />

The Payoffs of a Persistent Patient<br />

H Alter<br />

8 New Insights into the HCV Replication Cycle: Lessons Learned from 04<br />

Novel Culture Systems<br />

R Bartenschlager<br />

8 HCV Cell Culture Systems and the Development of Antiviral Therapeutics 05<br />

TJ Liang<br />

9 Role of Negative Regulatory Signals in the Pathogenesis of HCV 06<br />

A Grakoui<br />

10 A Model System for Hepatitis B and C Virus Co-infection 07<br />

V Brass<br />

10 Insights into the Impact of HBV and HCV Viral Dynamics on Antiviral Therapy 08<br />

AS Perelson<br />

11 Interferon Modulation of Cellular MicroRNAs as a Novel Antiviral Mechanism 09<br />

IM Pedersen<br />

11 DNA Microarray Analysis of the NS3 and NS5B Genes from Hepatitis C 10<br />

Genotype 1a Infected Patients<br />

G Liu-Young<br />

12 Assessment of Both Virological Response at Week 4 and at Week 12 11<br />

Optimizes Prediction of Treatment Outcome in Patients with Chronic<br />

Hepatitis C Treated with Peginterferon Alfa-2b Plus Ribavirin<br />

M Martinot-Peignoux<br />

13 Week 24 is the Optimal Time Point for Predicting Outcomes at 2 Years 12<br />

with Telbivudine<br />

KR Reddy<br />

14 Green Fluorescence Based Assays for Hepatitis C Virus Replication and 13<br />

Infectivity in Cell Culture<br />

S Dash<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xvii


page session title and author abstract<br />

15 Development of Mouse Model for Hepatitis C Virus Replication 14<br />

S Dash<br />

16 A Cell-Based, Miniaturized Infection System to Identify Bioactive Molecules 15<br />

Against Hepatitis C Virus<br />

P Gastaminza<br />

16 Generation of HCV E2 Specific Neutralizing Monoclonal Antibody 16<br />

SJ Park<br />

17 A Mutation within the Hepatitis C Virus NS3 Helicase Domain Promotes 17<br />

Virion Assembly in Cultured Hepatoma Cells<br />

M Yi<br />

19 Diagnosis and New Treatments for HBV and HCV<br />

21 HCV Replicons vs. Infectious Virus Systems in Drug Discovery 18<br />

SM Lemon<br />

21 The Size of the Viral Inoculum Determines the Kinetics, Magnitude and 19<br />

Outcome of Hepatitis B Virus Infection<br />

FV Chisari<br />

22 Population Genetics of HBV and HCV Quasispecies: Implications for Drug 20<br />

Development and Drug-Resistance Testing<br />

RW Shafer<br />

23 Nitazoxanide: A Potent Antiviral Agent Against HCV and HBV 21<br />

BE Korba<br />

24 Clinical Implications of Combined Intra-Cellular and Cellular Evolution of 22<br />

HCV Resistance during Direct Anti-HCV Treatment<br />

AU Neumann<br />

25 Phase 2 Studies with Albinterferon alfa-2b (alb-IFN) Dosed q4wk Provide 23<br />

Insights into Dose Selection for Future Studies<br />

I Jacobson<br />

25 Robust Antiviral Efficacy of a “Finger-loop” Allosteric Inhibitor of the HCV 24<br />

Polymerase in HCV Infected Chimpanzees<br />

R De Francesco<br />

26 Preclinical Development of the Amphipathic DNA Polymer REP 9AC for the 25<br />

Treatment of HBV Infection<br />

A Vaillant<br />

27 Inhibition of Hepatitis C Virus Replication by Octadecyloxypropyl -(S)-HPMPA 26<br />

in Genotype 1B and 2A Replicons<br />

KY Hostetler<br />

xviii Global Antiviral Journal Volume 3, Supplement 2


page session title and author abstract<br />

28 FKBP8 Plays a Crucial Role in the Replication of Hepatitis C Virus 27<br />

Y Matsuura<br />

29 Initial Recommendations for HCV Drug Resistance Analysis: A Consensus 28<br />

Statement from the HCV Drug Resistance Advisory Group<br />

K Lin<br />

30 Early Biochemical and Virological Response of Clevudine Therapy in Patients 29<br />

with HBV Associated Liver Cirrhosis<br />

KW Chung<br />

30 Clevudine was Superior to Lamivudine in the Patients with HBeAg(+) 30<br />

Chronic Hepatitis B<br />

GK Lau<br />

31 Clevudine Monotherapy Showed Rapid Viral and Biochemical Response in 31<br />

Chronic Hepatitis B Patients with Cirrhosis<br />

CH Lee<br />

32 Phase I Evaluation of a Novel Oral HCVp7 Inhibitor, BIT225, in Healthy 32<br />

Volunteers<br />

CA Luscombe<br />

32 Novel Mechanism of Action of Clevudine Triphosphate: Evidence for 33<br />

Non-competitive Inhibition of Hepatitis B Virus DNA Polymerase<br />

E Murakami<br />

33 Mechanistic Characterization of Potent Small Molecule HCV Inhibitors that 34<br />

Target NS5A<br />

A Sandrasagra<br />

34 GSK949614; a Novel and Potent Thumb Site Inhibitor of the HCV NS5B 35<br />

Polymerase<br />

PA Thommes<br />

35 Pyrrolo[1,2-b]pyridazin-2-ones Show Promising In Vitro Antiviral Activity 36<br />

Against HCV NS5B Polymerase<br />

CV Tran<br />

36 4-(1,1-Dioxo-1,4-dihydro-1λ 6 -benzo[1,4]thiazin-3-yl)-5-hydroxy-2H- 37<br />

pyridazin-3-ones are a Novel Series of Molecules which Inhibit the<br />

HCV NS5B Polymerase<br />

CV Tran<br />

37 Interference of Hsp90 Activity by Hsp90 Inhibitor Suppresses Hepatitis 38<br />

C Virus (HCV) Replication<br />

S Ujino<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xix


page session title and author abstract<br />

39 Hepatitis B Foundation Symposium:<br />

Early Detection and Management of Cirrhosis and Hepatocellular Carcinoma<br />

41 Hepatocellular Carcinoma: Why Early Diagnosis is Needed 39<br />

AM Di Bisceglie<br />

41 Assessment of Fibrosis: Non Biopsy Methods to Determine Hepatic Fibrosis 40<br />

NH Afdhal<br />

43 Screening Fibrosis: “La Révolution Française” 41<br />

T Poynard<br />

44 Treatment of HCC Over the Past Decade: The Experience of Over Four 42<br />

Thousand Patients in Japan<br />

M Omata<br />

45 Use of Pre-emptive Nucleoside Analogue Therapy for Hepatitis B Reactivation 43<br />

after Chemotherapy<br />

GKK Lau<br />

47 Immunology & Pathogenesis<br />

49 Cellular Immune Responses Against Hepatitis C in Acute and Chronic Infection 44<br />

MJ Koziel<br />

49 Broadly Neutralizing Antibodies to HCV: Definition of Epitopes and 45<br />

Antiviral Activity In Vitro and In Vivo<br />

M Law<br />

50 Innate Immune Studies in Hepatitis B: Novel Studies in Pathogenesis 46<br />

and Treatment<br />

K Visvanathan<br />

51 Early Steps in the Replicative Cycle of Hepatitis B Virus 47<br />

D Glebe<br />

52 Therapeutic Vaccination Against Chronic Hepatitis C Virus Infection: 48<br />

Towards a Proof-of-concept in Man?<br />

CS Klade<br />

53 Amelioration of Metabolic Disturbances and Oxidative Stress in 49<br />

Hepatitis C Viral Infection by FK506 (Tacrolimus)<br />

K Koike<br />

53 Poor Costimulatory Effects of CD 137 in Patients with Hepatocellular 50<br />

Carcinoma<br />

JW Shin<br />

54 Studies on a Therapeutic Vaccine by Modulating Host Dentritic Cells 51<br />

in Viral Hepatitis B Patients<br />

YM Wen<br />

xx Global Antiviral Journal Volume 3, Supplement 2


page session title and author abstract<br />

55 HBsAg-HBIg Complex Modulates Antigen Presentation of Dendritic Cells 52<br />

from Chronic Hepatitis B Patients<br />

B Zheng<br />

57 New Therapeutic Approaches<br />

59 HCV Entry Pathways and Implications for the Development of 53<br />

Entry Inhibitors<br />

MJ Evans<br />

59 The Role of CD81 and Scavenger Receptor Class B Member I in HCV Entry 54<br />

P Balfe<br />

60 Lipid Droplet is an Important Organelle for Production of Infectious 55<br />

Hepatitis C Virus<br />

K Shimotohno<br />

61 The Role of Cyclophilins and Cyclophilin Inhibitors in the Replication of HCV 56<br />

R Crabbé<br />

62 HCV Infection Increases Claudin-1 and Claudin-7 Expression by Inducing 57<br />

Cirrhosis<br />

A Kiss<br />

63 Development of Novel Hyperglycosylated Type 1 Interferons: A Strategy to 58<br />

Improve PK Performance Without Loss of Biological Potency<br />

LM Blatt<br />

64 Bioinformatics Resources Supporting the Analysis of Hepatitis C Virus 59<br />

EJ Lefkowitz<br />

65 A Combination of Direct Antiviral Compounds Can Achieve Sustained Viral 60<br />

Response in Hepatitis C Virus-Infected Chimpanzees<br />

DB Olsen<br />

65 Potent Antiviral Activity of the Nucleoside HCV Inhibitor, R7128, in Prior 61<br />

IFN Non-responders<br />

JG McHutchison<br />

66 R1626 Demonstrates Synergistic Antiviral Effect in Combination with 62<br />

Peginterferon Alfa-2a [40KD], with or without Ribavirin, and High Barrier<br />

to Resistance Development<br />

D Nelson<br />

67 Tenofovir DF (TDF) Showed Superior Antiviral Efficacy to Adefovir 63<br />

Dipivoxil (ADV) at 48 Weeks in Two Pivotal, Randomized, Double-Blind,<br />

Studies for the Treatment of HBeAg-Negative (-) and HBeAg-Positive (+)<br />

Chronic Hepatitis B (CHB): Study 102 and Study 103<br />

P Marcellin<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xxi


page session title and author abstract<br />

68 Sequence Sections of the 5‘-Non-Coding Region of the Hepatitis C-Virus 64<br />

RNA Genome at the DNA Level: Their Diverse Forms of Composition<br />

RH Dennin<br />

69 A Novel Anti-viral Compound, BIT225, Inhibits Bovine Diarrhea Virus 65<br />

(BVDV) In Vitro, and has Synergistic Antiviral Activity in Combination with<br />

Recombinant Interferon Alpha-2b (rIFNα−2b) and Ribavirin<br />

G Ewart<br />

70 Identification of Therapeutic Targets in Hepatitis B Virus (HBV) Associated 66<br />

Hepatocellular Carcinoma (HCC)<br />

MA Feitelson<br />

71 A Phase 1, Randomized, Blinded, Placebo-controlled, Single-dose, Dose- 67<br />

escalation Study of PEG-Interferon lambda (PEG-rIL-29) in Healthy Subjects<br />

DF Hausman<br />

72 Influence of Laboratory Parameters on Appearance and Course of Bleeding in 68<br />

Patients with Portal Hypertension and Liver Cirrhosis<br />

L Husova<br />

73 Structural Studies of the Hepatitis C Virus NS5A Protein 69<br />

R Love<br />

73 Dose Selection of Albinterferon Alfa-2b (alb-IFN) for a Phase 3 Clinical 70<br />

Program<br />

JG McHutchison<br />

74 Efficacy of Cationic Lipid-DNA Complexes (CLDC) on Hepatitis B Virus in 71<br />

Transgenic Mice<br />

JD Morrey<br />

75 Role of Stomatin in the Assembly of Hepatitis C Virus RNA Replicase 72<br />

Complex<br />

J-W Oh<br />

76 Antiviral Activity of Amino Acid Derivatives of Monascus Pigment in 73<br />

Hepatitis C Virus Replicating Cells<br />

J-W Oh<br />

77 Silymarin Displays Anti-viral, Anti-inflammatory, and Immunomodulatory 74<br />

Effects Towards Hepatitis C Virus<br />

S Polyak<br />

77 Construction and Applications of a Liver-specific Lentivirus Vector with 75<br />

Host Range Determined by the Envelope Proteins of HBV<br />

J Taylor<br />

xxii Global Antiviral Journal Volume 3, Supplement 2


page session title and author abstract<br />

78 Designed Zinc Finger Proteins Bind Duck Hepatitis B Virus Enhancer 76<br />

DNA and Decrease Production of Viral RNA, Proteins and Progeny In Vitro<br />

KA Zimmerman<br />

81 Resistance to Antiviral Agents<br />

83 Overcoming HCV Treatment-resistant Characteristics 77<br />

MW Fried<br />

84 Genetic and Structural Variability of the Hepatitis C Viral Polymerase, NS5B: 78<br />

Implications for Resistance to Inhibitors<br />

PC Simister<br />

85 Selection of Multidrug Resistant Hepatitis B Virus Following Sequential 79<br />

Monotherapy and Combination Therapy<br />

A Ayres<br />

86 Multidrug Resistance and Cross-resistance Pathways in HBV as a 80<br />

Consequence of Treatment Failure<br />

A Ayres<br />

87 Antiviral Effect of Nucleoside Analogs and Interferon on a Novel 81<br />

Infectious Cloned Hepatitis C Virus Containing the S282T Mutation in the<br />

NS5B RNA Polymerase<br />

L Bassit<br />

88 Understanding the Molecular Basis of HBV Drug Resistance by Molecular 82<br />

Modeling<br />

CK Chu<br />

89 High Genetic Barrier to HCV Resistance Presented by PSI-6130 83<br />

A De La Rosa<br />

90 Determination of Lamivudine, Adefovir and Entecavir Resistance in Acute 84<br />

and Chronic Hepatitis B Virus Infections<br />

C Eroglu<br />

90 Databases for Antiviral Resistance Monitoring in Hepatitis B 85<br />

S Locarnini<br />

91 Combination Chemotherapy with Nucleos(t)ide Analogue Reverse 86<br />

Transcriptase Inhibitors (NRTI) May Suppress Multidrug-resistant HBV:<br />

Using In Vitro Assays to Identify Optimal NRTI Combinations and Doses<br />

T Sozzi<br />

92 Relative Replication Capacity and Antiviral Cross-Resistance Phenotypes 87<br />

of Multidrug-resistant Hepatitis B Virus Mutants Implicated in<br />

Non-response to Sequential Treatment with Adefovir, Lamivudine<br />

and Entecavir<br />

T Sozzi<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xxiii


page session title and author abstract<br />

95 Co-infection with HIV and Other Viruses<br />

97 Treatment of HBV/HIV Co-infections 88<br />

MG Peters<br />

97 Treatment of HCV in HIV-infected Individuals 89<br />

Z Temesgen<br />

98 HCV/HIV Co-infections: Challenges from the Perspective of TAG 90<br />

T Swan<br />

99 Tolerability of Darunavir/Ritonavir Versus Lopinavir/Ritonavir in 91<br />

Lopinavir/Ritonavir-naïve, Treatment-experienced, Hepatitis B or C<br />

Co-infected Patients in TITAN<br />

YK Dayaram<br />

100 The Effect of Lopinavir/Ritonavir on HIV/HCV Co-Infected Individuals 92<br />

CA Smith<br />

100 Pharmacokinetics of TMC125 in HIV-negative Volunteers with Mild or 93<br />

Moderate Hepatic Impairment<br />

F Tomaka<br />

101 Tolerability of Once-daily Darunavir/r versus Lopinavir/r in 94<br />

Treatment-naïve, Patients Co-infected with Hepatitis B and/or C in the<br />

ARTEMIS Trial<br />

F Tomaka<br />

102 Pharmacokinetics of Multiple-Dose Darunavir in Combination with 95<br />

Low-Dose Ritonavir in Individuals with Impaired Hepatic Function<br />

F Tomaka<br />

103 TMC125 Safety and Tolerability in Treatment-Experienced Hepatitis B 96<br />

or C Co-infected Patients in DUET-1 and DUET-2<br />

F Tomaka<br />

105 Pharmacology and Drug Interactions of HBV and HCV Therapeutics<br />

107 Preclinical Drug Metabolism in Viral Hepatitis Drug Discovery 97<br />

AS Ray<br />

107 Frontiers of Clinical Pharmacology in Hepatitis Drug Development 98<br />

CW Flexner<br />

109 Current and Future: HCV Maintenance Therapy 99<br />

KL Lindsay<br />

110 The Role of Pgp-driven Efflux on the Potency of HCV Replication Inhibitors 100<br />

F McPhee<br />

xxiv Global Antiviral Journal Volume 3, Supplement 2


page session title and author abstract<br />

111 Pharmacologic Evaluation of Novel Small Molecule HCV Inhibitors 101<br />

Affecting NS5A-dependent Functions<br />

CR Wobbe<br />

113 Optimizing the Outcome of Therapeutics for HBV and HCV<br />

115 Measuring and Mapping Partnership Integration for Optimizing 102<br />

Assessment and Treatment of Hepatitis C<br />

G Butt<br />

115 Development of a Novel Mouse Model to Evaluate Single and 103<br />

Combination Therapy Against Hepatitis B Virus<br />

MA Feitelson<br />

116 Association of Pretreatment Serum Interferon-γ-inducible Protein 10 104<br />

(Ip-10) Levels with Sustained Virological Response to Peginterferon Plus<br />

Ribavirin Therapy for Chronic Hepatitis C (CHC) with Virus Genotype 1<br />

Infection<br />

M Fukuda<br />

117 Optimizing Uptake and Response to Treatment of Hepatitis C Virus 105<br />

(HCV) Infection in Injection Drug Users (IDUs): A Novel Model<br />

Incorporating Multidisciplinary Care, Directly Observed Therapy and<br />

Peer-support<br />

J Grebely<br />

118 Efficacy of Pegylated Interferon Alpha-2a and Ribavirin Treatment in 106<br />

Chronic Hepatitis C Patients Depends on Various Baseline Parameters<br />

and Early Viral Kinetics<br />

P Husa<br />

119 Peginterferon Alpha-2a and Ribavirin Combination Therapy for Chronic 107<br />

Hepatitis C in Patients with Hemophilia – Preliminary Report<br />

HJ Kim<br />

120 Changes of IFN-inducible Gene (IP-10, PKR, MxA) Expressions During 108<br />

Pegylated Interferon Alfa-2b Plus Ribavirin Therapy in HCV Genotype 1<br />

Infected Japanese Patients<br />

R Nakao<br />

121 Baseline Characteristics and Early Virologic Response to Telbivudine Predict 109<br />

2‐Year Outcomes for Patients with HBeAg-negative Chronic Hepatitis B<br />

T Poynard<br />

122 Predictors of Sustained Viral Response in the Retreatment of Previous 110<br />

Interferon/Ribavirin Nonresponders: Results from the EPIC 3 Program<br />

T Poynard<br />

123 Addition of Lamivudine to Patients with Chronic Hepatitis B who are 111<br />

Viremic on Adefovir Dipivoxil Lowers Plasma HBV DNA Levels<br />

J Ryan<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis<br />

xxv


page session title and author abstract<br />

123 A Systematic Review of the Effectiveness of Pegylated Interferon, 112<br />

Lamivudine, Adefovir and Entecavir for the Treatment of Hepatitis B<br />

G Woo<br />

124 Safety Recommendations for Laboratory Values in Specific Product 113<br />

Characteristics (SPC) of Peginterferon Alfa-2a (PEG) and Ribavirin (RBV) -<br />

What Happens Under Real Life Conditions?<br />

E Zehnter<br />

125 Standard of Medical Care for Patients with Chronic Hepatitis C (cHC) and 114<br />

Liver Cirrhosis in Germany - A Status Report<br />

E Zehnter<br />

127 Late Breaker Session<br />

129 Long Term Follow-Up of HCV Patients Completing Peg-IFN Plus Ribavirin 115<br />

Treatment – Is There a Cure?<br />

MP Manns<br />

130 Delta Hepatitis: Treatment Options for a Largely Neglected Disease 116<br />

MP Manns<br />

130 Biochemical Mechanism of Entecavir as an Antiviral Polymerase Inhibitor 117<br />

KS Anderson<br />

131 Advancing RNAi-based Therapeutic from Discovery to Clinic 118<br />

C Pachuk and R Gish<br />

131 Ultra-deep Pyrosequencing of HBV Quasispecies in Nucleoside Analog 119<br />

Treated and Untreated Patients<br />

S Margeridon-Thermet<br />

xxvi Global Antiviral Journal Volume 3, Supplement 2


State of the Art Lectures<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 1


ABSTRACT 01<br />

The Changing Global<br />

Epidemiology of HBV and HCV<br />

K Fenton<br />

Centers for Disease Control and Prevention, Atlanta, USA<br />

Hepatitis B Virus (HBV) and Hepatitis C Virus (HCV)<br />

infections continue to have a severe and pervasive<br />

impact on the health of millions around the world.<br />

Today, an estimated 350 million persons are living<br />

with chronic HBV and there are currently 500-700,000<br />

HBV-related deaths/year. There are an estimated<br />

120-180 million individuals living with chronic HCV<br />

with approximately 370,000 HCV-related deaths/<br />

year. Both infections have a disproportionate<br />

and increasing burden in developing countries.<br />

Transmission modes for HBV include perinatal,<br />

parenteral, sexual transmission; and persons infected<br />

as newborns and young children have highest risk<br />

of chronic disease. For HCV, healthcare exposures<br />

are particularly problematic in developing countries,<br />

whereas injecting drug use are among the major<br />

determinants in developed country settings.<br />

Preventing HBV and HCV in the United States and<br />

globally remains challenging. Despite some success of<br />

infant vaccination programs - by 2005, 80% of WHO<br />

Member States had introduced HBV vaccination<br />

programs and an estimated 55% of the world's<br />

children less than 1 year of age had received 3 doses<br />

of HepB – much remains to be done as coverage varies<br />

by WHO region. However, progress is being observed<br />

as a result of global advocacy, decreasing vaccine<br />

prices, and availability of resources to the poorest<br />

countries. Vaccinating high risk adults is also a major<br />

public health priority and barriers to adult HBV<br />

vaccination include: Fiscal concerns; cost of vaccine;<br />

reimbursement for supply and delivery; provider<br />

practices; time constraints; and patient acceptance.<br />

For HCV, prevention strategies include protecting<br />

the blood and tissue supply; promoting safe injection<br />

practices; assuring infection control in health care<br />

settings; promoting HCV screening for persons at<br />

risk; and referring for care and treatment to stop<br />

liver disease and transmission. However the global<br />

challenges to prevention include the high incidence<br />

of infection among IDUs; the tremendous unmet<br />

need for HCV counseling and harm reduction; higher<br />

prevalence of infection; lower rate of viral clearance;<br />

lower rates of response to treatment; greater risk of<br />

liver cancer.<br />

This presentation will provide an overview of the<br />

evolving global epidemiology of HBV and HCV<br />

infections; current prevention and control strategies;<br />

and reflect upon recent developments and future<br />

plans related to preventing HBV and HCV in the<br />

United States.<br />

ABSTRACT 02<br />

New Insights into Hepatitis B<br />

Pathogenesis and Resistance<br />

S Locarnini<br />

Head, R&MD, VIDRL and Director, WHO Collaborating<br />

Centre for Virus Reference and Research, Melbourne,<br />

Victoria, 3051, Australia<br />

The hepatitis B virus (HBV) has developed a number of<br />

strategies to ensure its persistence in the infected host.<br />

The production of excess hepatitis B surface antigen<br />

(HBsAg), the expression of hepatitis Be antigen<br />

(HBeAg) and the use of a viral minichromosome (in<br />

which can be found the covalently closed circular [ccc]<br />

DNA), a transcriptional template which guarantees<br />

virus production with minimal interference from<br />

the host’s immune response. The pathogenesis of<br />

hepatitis B is the outcome of the interplay between<br />

HBV, the hepatocyte and the immune response,<br />

since under normal circumstances, the virus is not<br />

cytopathic. The liver damage of chronic hepatitis<br />

B (CHB) is the result of the host’s cellular immune<br />

response to HBV-infected hepatocytes as part of the<br />

“immune clearance” phase of the disease. Thus, the<br />

clinical presentations and natural history of CHB are<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 3


mediated though complex interactions between the<br />

virus and host immune response. Both the innate and<br />

adaptive branches of the host immune response need<br />

to be considered as a coordinated and dynamic line of<br />

attack, rather than as consecutive and independent<br />

entities. Both established and potential roles for<br />

many components of the innate immune system have<br />

been identified. In particular, important interactions<br />

between HBeAg, HBV and Toll-like receptors (TLR-2),<br />

Kupffer cells, natural killer T-cells, and dendritic<br />

cells have been described (see Visvanathan, K. et al<br />

2007 Hepatology;45:102). Achieving clearance of<br />

HBV appears to require initial viral suppression and<br />

recruitment of effector cells by the innate immune<br />

system, along with adequate antigen presentation<br />

to and activation of the adaptive immune response<br />

arm. With the increasing challenge of antiviral drug<br />

resistance to nucleos(t)ide analogues, a greater<br />

understanding of the immunological mechanisms<br />

involved in CHB has the potential to identify new<br />

therapeutic targets in order to eventually control this<br />

challenging viral infection of man.<br />

4 Global Antiviral Journal Volume 3, Supplement 2


Advances in New Models<br />

for HBV and HCV<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 5


Abstract 03<br />

A Pre-mortem Lookback<br />

Investigation of One's Life<br />

in Research: The Payoffs of a<br />

Persistent Patient<br />

H Alter<br />

National Institutes of Health, USA<br />

Harvey J Alter, MD, serves as Chief, Infectious<br />

Disease Section and Associate Director for Research,<br />

Department of Transfusion Medicine, Clinical Center,<br />

NIH. Dr. Alter was coinvestigator in the discovery<br />

of the Australia antigen and principal investigator<br />

in the first study to biophysically characterize that<br />

antigen. He was principal investigator in prospective<br />

studies that identified the clinical entity non-A,<br />

non-B (NANB) hepatitis and was the first to prove<br />

NANB was a transmissible agent and to establish the<br />

chimpanzee model for study of this disease. Dr. Alter<br />

collaborated in a series of studies that defined the<br />

natural history of NANB/HCV infection and proved<br />

its frequent progression to chronic hepatitis and<br />

cirrhosis, a prophetic concept at that time.<br />

Dr. Alter was principal investigator in sequential<br />

prospective studies of transfusion-associated<br />

hepatitis that established the inordinate risk of paid<br />

donor and HBsAg positive blood, that demonstrated<br />

the risk of ALT and anti-HBc positive blood, and that<br />

were instrumental in influencing a national blood<br />

policy that mandated HBsAg and surrogate assay<br />

screening. These unique prospective studies have<br />

documented the progressive decline of TAH incidence<br />

from 33% in the 1960s to near zero in 1997 and have<br />

sequentially established the efficacy of various donor<br />

screening interventions.<br />

studies that defined the size, buoyant density and<br />

infectivity titer of the NANB/HCV agent, that defined<br />

replication characteristics and mutation rates, and<br />

that characterized neutralizing antibody responses.<br />

Dr. Alter was principal investigator in a large cohort<br />

study to define the modes of transmission and clinical<br />

relevance of asymptomatic HCV infection and first to<br />

suggest HCV may be spread by cocaine snorting. He<br />

was senior investigator in a study that characterized<br />

the HCV quasispecies early in infection and related<br />

the complexity and diversity of the quasispecies to<br />

the severity of clinical outcome.<br />

For these studies, Dr. Alter has been awarded the<br />

DHEW Superior Service Award and Distinguished<br />

Service Medal, the latter the highest award conferred<br />

to persons in the Public Health Service. Dr. Alter<br />

was corecipient of the Landsteiner Prize, the most<br />

prestigious award of the American Association<br />

of Blood Banks. For his cumulative research<br />

accomplishments, Dr. Alter was elected to fellowship<br />

in the American Association of Physicians and is the<br />

year 2000 recipient of the Clinical Lasker Award. In<br />

2001, Dr. Alter was elected to the National Academy<br />

of Sciences and in 2002 to the Institute of Medicine.<br />

He was also elected to Master status in the American<br />

College of Physicians. In 2004, he was recipient of<br />

the First International Medal for Science given by<br />

INSERM, the French counterpart to NIH and recipient<br />

of the American College of Physicians Award for<br />

Outstanding Work in Science as Related to Medicine.<br />

Following the cloning of HCV, Dr. Alter conducted the<br />

key study that established HCV as the major cause<br />

of transfusion-associated hepatitis and proved the<br />

clinical efficacy of anti-HCV screening assays. He was<br />

a collaborator in a series of human and chimpanzee<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 7


Abstract 04<br />

New Insights into the HCV<br />

Replication Cycle: Lessons<br />

Learned from Novel Culture<br />

Systems<br />

R Bartenschlager<br />

Department for Molecular Virology, University of<br />

Heidelberg, Im Neuenheimer Feld 345, Heidelberg,<br />

Germany<br />

Hepatitis C viruses (HCV) comprise a group of<br />

positive-strand RNA viruses that together with<br />

the flaviviruses and the pestiviruses belong to the<br />

Flaviviridae family. As a major cause of acute and<br />

chronic liver disease worldwide for which no selective<br />

therapy exists, HCV has received much attention.<br />

The HCV genome encodes a single polyprotein that<br />

is cleaved into 10 different products. To most of<br />

these proteins distinct functions could be ascribed<br />

and the 3D X-ray crystal structures have been solved<br />

for several viral enzymes that are prime targets for<br />

antiviral therapy, especially the NS3 protease and the<br />

NS5B RNA-dependent RNA polymerase (RdRp).<br />

With the availability of cell culture model systems,<br />

in particular HCV replicons and the HCVcc system,<br />

new insights into the molecular and cellular<br />

mechanisms underlying HCV entry, replication,<br />

assembly and egress have been gained. For instance,<br />

novel molecules involved in viral entry have been<br />

identified and a complex picture emerges how the<br />

virus productively infects a cell. Likewise, new and<br />

surprising insights have been gained how HCV<br />

particles assemble. It turned out that lipid droplets<br />

play a very important role for virion morphogenesis,<br />

and that NS5A appears to be a key regulator of the<br />

different steps of the viral replication cycle. Finally,<br />

high-throughput screening methods have been used<br />

to identify host cell factors that contribute to HCV<br />

replication. Prominent examples are cyclophilins that<br />

appear to activate NS5B RdRp activity, and h-VAPA as<br />

well as FBL-2 interacting with NS5A and contributing<br />

to RNA replication. The common denominator is that<br />

HCV usurps multiple cellular processes, including<br />

lipid metabolism to achieve efficient RNA replication,<br />

virus assembly and virion infectivity.<br />

Abstract 05<br />

HCV Cell Culture Systems and<br />

the Development of Antiviral<br />

Therapeutics<br />

TJ Liang<br />

Liver Diseases Branch, NIDDK, NIH, USA<br />

Hepatitis C virus (HCV) is a leading cause of morbidity<br />

and mortality worldwide. The pathogenesis of hepatitis<br />

C and effects of HCV gene expression on infected cells<br />

remain unclear in vivo. Efforts to establish cell culture<br />

and animal models for hepatitis C are critical for the<br />

understanding of the virus and the disease it causes,<br />

as well as for developing antiviral therapeutics.<br />

We recently showed the production of HCV particles<br />

by using a DNA expression plasmid containing fulllength<br />

HCV cDNA flanked by self-cleaving ribozymes.<br />

This construct also contains the secreted alkaline<br />

phosphatase gene to monitor the expression from this<br />

construct and to normalize transfection efficiency<br />

and to control for effects of culture conditions, such<br />

as anti-viral testing. We produced HCV particles of<br />

various genotypes including 1a (H77), 1b (CG1b)<br />

and 2a (J6 and JFH-1) in the HCV-ribozyme system.<br />

The constructs also contain the secreted alkaline<br />

phosphatase gene to control for transfection<br />

efficiency and effects of culture conditions. After<br />

transfection into Huh7-derived cell line Huh7.5.1,<br />

continuous HCV replication and secretion were<br />

confirmed by detection of HCV RNA and core antigen<br />

in the culture medium. HCV replication levels of<br />

strains H77, CG1b and J6 were comparable, whereas<br />

the JFH-1 strain replicates at a substantially higher<br />

level than the other strains. Interferon-alfa treatment<br />

8 Global Antiviral Journal Volume 3, Supplement 2


can substantially supporess HCV replication in this<br />

model. To evaluate the infectivity in vitro, the culture<br />

medium of JFH-1-transfected cells were inoculated<br />

to naïve Huh7.5.1 cells. HCV proteins were detected<br />

by immunofluorescence 3 days after inoculation. To<br />

evaluate the infectivity in vivo, the culture medium<br />

from HCV genotype 1b-transfected cells was<br />

inoculated into a chimpanzee and caused a typical<br />

course of HCV infection. The HCV 1b propagated in<br />

vitro and in vivo had identical sequences as the HCV<br />

genomic cDNA used for cell culture transfection.<br />

Current treatment of chronic hepatitis C based<br />

on combination of peginterferon and ribavirin is<br />

only effective in about half of the patients and is<br />

accompanied by substantial side effects. Developing<br />

new classes of drugs against HCV is crucial. A new class<br />

of HCV inhibitors (amphipathic DNA polymers) that<br />

are active in the viral entry step will be presented. Its<br />

potential mechanism of action and value in dissecting<br />

the molecular pathway of HCV entry will be discussed.<br />

The development of culture systems for production<br />

of various HCV genotypes provides a valuable tool<br />

not only to study the replication and pathogenesis of<br />

HCV but also to screen for antivirals.<br />

Abstract 06<br />

Role of Negative Regulatory Signals<br />

in the Pathogenesis of HCV<br />

A Grakoui<br />

Emory Vaccine Center, USA<br />

The adaptive immune response relies upon controlled<br />

activation of antigen specific T lymphocytes which<br />

are critical for maintaining immunity to pathogens<br />

as well as tolerance to host tissues. The recently<br />

described PD-1/PD-L1 pathway is important for both<br />

responsibilities of the adaptive immune response.<br />

PD-1 (programmed death 1) is a co-inhibitory receptor<br />

expressed by T cells whose engagement by its ligands<br />

PD-L1 and PD-L2 results in decreased proliferation<br />

and cytokine production by antigen specific T cells.<br />

Studies in the most well established murine model<br />

of chronic infection, lymphochoriomeningitis virus<br />

(LCMV), first demonstrated the exciting possibility<br />

that functional blockade of the PD-1/PD-L1 pathway<br />

could reverse functionally impaired antigen specific<br />

T cells. And indeed, subsequent studies in HIV<br />

showed that PD-1 expression on HIV-specific CD8 + T<br />

cells was upregulated and that PD-1 expression levels<br />

correlated both with impaired capacity for cytokine<br />

production and proliferation as well as viral load.<br />

We hypothesized that this important co-inhibitory<br />

pathway may contribute to hepatitis C virus (HCV)<br />

persistence in humans and established a large patient<br />

cohort in which to study the negative regulation of<br />

the anti-HCV immune response. Since there is no<br />

vaccine available to prevent HCV infection and current<br />

treatment regimens yield a sustained viral response<br />

of less than 50%, elucidating the points at which<br />

host immunity fails to abrogate viremia is critical for<br />

the development of immune interventions that may<br />

augment the host response to HCV. To this end, we<br />

have found that HCV specific CD8 + T cells found in<br />

the peripheral blood of patients with persistent HCV<br />

infection express high levels of the co-inhibitory<br />

receptor, PD-1. These cells have impaired proliferative<br />

capacity that can be reversed by PD-1/PD-L1 blockade.<br />

Importantly, intrahepatic HCV-specific cells not only<br />

express high levels of PD-1, but also low levels of<br />

the IL-7 receptor (CD127), characteristic findings of<br />

functionally exhausted T cells. These phenotypically<br />

exhausted cells are compartmentalized to the liver,<br />

the site of active viral replication, suggesting that<br />

repeated exposure to viral antigen may contribute to<br />

regulation of PD-1 expression. The functional reversal<br />

of T cell exhaustion by PD-1/PD-L1 blockade holds<br />

promise for a possible new therapeutic intervention<br />

that will revive dysfunctional T cells in persistent<br />

viral infection.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 9


Abstract 07<br />

A Model System for Hepatitis B<br />

and C Virus Co-infection<br />

P Bellecave 1 , J Gouttenoire 1 , M Gajer 2 , V Brass 2 ,<br />

G Koutsoudakis 3 , HE Blum 2 , M Nassal 2 ,<br />

R Bartenschlager 3 , and D Moradpour 1<br />

1 Division of Gastroenterology and Hepatology, Centre<br />

Hospitalier Universitaire Vaudois, University of Lausanne,<br />

CH-1011 Lausanne, Switzerland; 2 Department of<br />

Medicine II, University of Freiburg, D-79106 Freiburg,<br />

Germany; 3 Department of Molecular Virology, University<br />

of Heidelberg, D-69120 Heidelberg, Germany<br />

Co-infection with hepatitis B virus (HBV) and<br />

hepatitis C virus (HCV) has been associated with<br />

severe liver disease and frequent progression to<br />

liver cirrhosis and hepatocellular carcinoma. Clinical<br />

evidence suggests reciprocal replicative suppression<br />

of the two viruses (‘viral interference’). However, due<br />

to the lack of appropriate model systems virtually<br />

nothing is known about molecular interactions<br />

between HBV and HCV. On this background, the aim<br />

of this study was to develop a model system to study<br />

HBV and HCV co-infection.<br />

A tetracycline-regulated gene expression system was<br />

used to generate stable Huh-7 cell lines inducibly<br />

replicating HBV. These cell lines and control Huh-7<br />

cell lines inducibly expressing the green fluorescent<br />

protein (GFP) were transfected with selectable HCV<br />

replicons or infected with cell culture-derived HCV<br />

(HCVcc).<br />

Three successive transfection and selection steps<br />

allowed the establishment of Huh-7 cells inducibly<br />

replicating HBV and constitutively replicating<br />

subgenomic HCV RNA. In these cell lines, it is now<br />

possible to regulate the expression of HBV proteins,<br />

HBV genome replication, and infectious HBV particle<br />

formation by the concentration of tetracycline in<br />

the cell culture medium while HCV proteins are<br />

expressed and HCV RNA replicated in a constitutive<br />

fashion. In a series of proof-of-principle experiments,<br />

this system was used to assess the antiviral effects of<br />

interferon-alpha and of specific inhibitors of the HBV<br />

polymerase as well as the HCV serine protease and<br />

NS5B polymerase. These experiments demonstrated<br />

that HBV and HCV replication can be selectively<br />

inhibited in cell lines harboring both viruses and<br />

that the presence or absence of replicating HBV does<br />

not significantly alter HCV RNA replication or the<br />

response of HCV to interferon-alpha. Preliminary<br />

immunofluorescence and confocal laser scanning<br />

microscopy data do not show colocalization between<br />

HBV and HCV proteins. In addition, HBV replication<br />

does not significantly alter the characteristics of<br />

HCV replication complexes. These initial results<br />

demonstrate that HBV and HCV can replicate in<br />

the same cell. Studies using HCVcc to infect HBVinducible<br />

cell lines are currently in progress to validate<br />

and extend these findings.<br />

In conclusion, we have successfully established and<br />

are currently exploiting a novel model system to<br />

investigate interactions between HBV and HCV.<br />

Understanding such interactions at the molecular<br />

level should yield new insights into the pathogenesis<br />

and clinical management of HBV-HCV co-infection.<br />

Abstract 08<br />

Insights into the Impact of HBV<br />

and HCV Viral Dynamics on<br />

Antiviral Therapy<br />

AS Perelson<br />

Theoretical Biology and Biophysics, Los Alamos National<br />

Laboratory, Los Alamos, NM 87545, USA<br />

Much of the modeling of HCV and HBV infection<br />

and treatment has centered around a simple model<br />

introduced by Neumann et al. in 1998 to model the<br />

first 14`days of treatment of HCV infection with daily<br />

interferon. While this model has been very successful<br />

it makes a number of simplifying assumptions. Here<br />

I shall show how the Neumann et al model has been<br />

improved so as to be able to incorporate proliferation<br />

of both uninfected and infected hepatocytes as<br />

10 Global Antiviral Journal Volume 3, Supplement 2


well time-varying changes in drug efficacy that is<br />

particularly relevant to once-weekly treatment with<br />

pegylated interferon. Using these new versions of<br />

the model I shall show how a variety of patterns of<br />

viral decline under therapy that have been called<br />

biphasic, triphasic, and stepwise declines as well as<br />

viral rebound can be explained.<br />

that IFNβ treatment leads to a significant reduction<br />

in the expression of the liver-specific miR-122, a<br />

miR that has been previously shown to be essential<br />

for HCV replication. Therefore, our findings strongly<br />

support the notion that mammalian organisms too,<br />

via the interferon system, utilize cellular miRs to<br />

combat viral infections.<br />

Abstract 09<br />

Interferon Modulation of Cellular<br />

MicroRNAs as a Novel Antiviral<br />

Mechanism<br />

IM Pedersen 1 , G Cheng 3 , S Wieland 3 , S Volonia 4 ,<br />

CM Croce 4 , FV Chisari 3 , and M David 1,2<br />

1 Department of Molecular Biology, Division of Biological<br />

Sciences; 2 Moores Cancer Center, University of California<br />

San Diego, La Jolla, CA 92093, USA; 3 Division of<br />

Experimental Pathology, The Scripps Research Institute,<br />

La Jolla, CA 92037, USA; 4 Department of Molecular<br />

Virology, Immunology & Medical Genetics, Ohio State<br />

University, Columbus, OH 43210, USA<br />

RNA interference through non-coding microRNAs<br />

(miRs) represents a vital component of the innate<br />

antiviral immune response in plants and invertebrate<br />

animals, however, a role for cellular miRs in the<br />

defense against viral infection in mammalian<br />

organisms has thus far remained elusive. We show<br />

now that interferon beta (IFNβ) rapidly modulates<br />

the expression of numerous cellular miRs, and<br />

that 8 of these IFNβ-induced miRs have sequencepredicted<br />

targets within the hepatitis C virus (HCV)<br />

genomic RNA. Introduction of synthetic miR-mimics<br />

corresponding to these IFNβ-induced miRs reproduces<br />

the antiviral effects of IFNβ on HCV replication and<br />

infection, whereas neutralization of these antiviral<br />

miRs with anti-miRs reduces the antiviral effects of<br />

IFNβ against HCV. Single nucleotide changes in the<br />

target seed sequences of miR-196 and miR-448 in the<br />

HCV genome render miRs-196 and -448 ineffective<br />

against HCV replication, however, introduction of<br />

compensatory nucleotide substitutions in the miRs<br />

restores their efficacy. Furthermore, we demonstrate<br />

Abstract 10<br />

DNA Microarray Analysis of<br />

the NS3 and NS5B Genes from<br />

Hepatitis C Genotype 1a Infected<br />

Patients<br />

G Liu-Young 1 , J Chiarella 1 , J Stapleton 2 ,<br />

G Garcia-Tsao 1 , J Grebely 3 , B Conway 3 , K Dieckhaus 4 ,<br />

PK Barua 1 and MJ Kozal 1<br />

1 Yale University and the VA CT Healthcare System,<br />

Connecticut, USA; 2 University of Iowa, Iowa, USA;<br />

3 University of British Columbia, Vancouver, BC, Canada;<br />

4 University of Connecticut, Connecticut, USA<br />

Background: The HCV coding region for new<br />

targets of HCV therapy encompass >5Kb (NS3,<br />

NS4A, NS5A and NS5B) of the viral genome, and<br />

investigations of drug resistance at one or more sites<br />

require extensive sequencing. The genetic diversity<br />

of major HCV genotypes further complicates assay<br />

development. New assays that can rapidly and<br />

accurately provide the sequence of target genes from<br />

major HCV genotypes could greatly facilitate drug<br />

resistance investigations.<br />

Methods: The entire HCV 1a genome and the NS3,<br />

NS4A, NS5A and NS5B genes from HCV genotypes<br />

1b, 2a, 2b, and 3a, have been placed on a customdesigned<br />

microarray. In addition, the microarray<br />

has arrays to detect minority variants containing<br />

drug resistance mutations in NS3, NS5A and NS5B<br />

for genotypes 1a and 1b. RT-PCR was performed<br />

to amplify the NS3 and NS5B genes from HCV 1ainfected<br />

patients. Amplicons were hybridized to the<br />

microarray and the sequences were analyzed to detect<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 11


polymorphisms/mutations. A consensus for 1a NS3<br />

and NS5B sequences was determined by analyzing<br />

data from HCV databases and the interrogated<br />

samples.<br />

Results: 129 unique HCV NS3 gene sequences from<br />

treatment naïve individuals (region of interest: codons<br />

32 to 173) were analyzed by microarray and standard<br />

sequencing and 56.8% of nucleotide (nt) and 42% of<br />

amino acid (aa) positions were polymorphic; of 109<br />

unique NS5B gene sequences (region of interest:<br />

codons 230 to 484) analyzed by microarray, 69.3%<br />

of nt and 29.8% of aa positions were polymorphic.<br />

Positions in NS3 gene associated with drug resistance<br />

to VX-950 and SCH503034 (codons 36, 54, 155, 156,<br />

168, and 170) and positions in NS5B associated with<br />

drug resistance to NM283 and HCV-796 (codons<br />

282 and 316) remained highly conserved and no<br />

resistance-associated aa changes were identified by<br />

microarray. The microarray determined the sequence<br />

of major HCV protease and polymerase inhibitor<br />

positions in 99.2% and 96.4% of samples respectively<br />

(range 95-100% depending on codon interrogated).<br />

Conclusions: The HCV microarray determined<br />

the sequence of a large portion of the HCV 1a genome<br />

in a single hybridization experiment, including the<br />

major NS3 protease and NS5B polymerase inhibitor<br />

resistance mutation sites. In this microarray survey<br />

of viral variants from HCV-infected therapy-naïve<br />

subjects, the NS3 and NS5B genes were highly<br />

polymorphic in nature. Further studies using specific<br />

arrays and Ultra Deep sequencing to detect very<br />

low levels of minor HCV variants will be required<br />

to determine if minor resistant variants pre-exist in<br />

patients’ samples.<br />

Abstract 11<br />

Assessment of Both Virological<br />

Response at Week 4 and at Week<br />

12 Optimizes Prediction of<br />

Treatment Outcome in Patients<br />

with Chronic Hepatitis C Treated<br />

with Peginterferon Alfa-2b Plus<br />

Ribavirin<br />

M Martinot-Peignoux, M-P Ripault, S Maylin,<br />

R Moucari, N Boyer, N Giuily, C Castelnau, and<br />

P Marcellin<br />

INSERM, U-773, Centre de Recherche Biomédicale<br />

Bichat-Beaujon CRB3; Université Paris VII, and Service<br />

d’Hépatologie, Hôpital Beaujon, Clichy, France<br />

Pretreatment viral load (VL) is an important predictor<br />

for treatment outcome in patients with chronic<br />

hepatitis C. Zeuzem et al. (AASLD 2006) proposed<br />

400 000 IU/ml as the optimal cut-off to best<br />

discriminate low and high VL, based on the<br />

probability to achieve SVR in patients treated with<br />

PEG-IFN+RBV.<br />

We aimed to analyze the positive predictive value<br />

(PPV) of this cut-off value at baseline, the PPV of<br />

rapid virological response at week 4 (RVR4) and the<br />

negative predictive value (NPV) of non early virological<br />

response at week 12 (non-EVR12), in patients treated<br />

with PEG-IFN alpha 2b+RBV.<br />

Patients-Methods: 408 patients (221 naïve;<br />

187 non-naïve) consecutively treated with standard<br />

schedule of PEG-IFN alpha-2b+RBV, were included.<br />

Serum HCV RNA was measured at baseline, week 4,<br />

week 12, end of treatment and 6 months after the<br />

end of treatment with the quantitative VERSANT R<br />

HCV 3.0 Assay (bDNA). Samples below the limit of<br />

quantification were tested with VERSANT R HCV RNA<br />

Qualitative Assay (TMA) (Siemens Medical Solution<br />

Diagnostic). SVR was defined as undetectable serum<br />

HCV RNA by TMA at end of 6 months post-treatment<br />

12 Global Antiviral Journal Volume 3, Supplement 2


follow-up. At treatment initiation PPV was defined<br />

with a cut-off set up at ≤ 400 10 3 IU/ml; at week 4<br />

the PPV was defined as TMA undetectable or ≥ 2 log<br />

drop baseline VL; at week 12 the NPV was defined<br />

as TMA detectable or < 2 log drop baseline viral<br />

load. A logistic regression analysis was performed<br />

to identify factors independently associated with<br />

RVR. The characteristics included were: baseline<br />

VL (≤vs>400x10 3 IU/ml), HCV genotype (1, 2, 3,<br />

4, 5), gender, age (≤vs>45y), histology grade (A0-<br />

1≤vs>A2-3), fibrosis stage (F0-2≤vs>F3-4) assessed<br />

with Metavir score, serum ALT, pretreatment status.<br />

Results: At baseline PPV of VL ≤ 400 000IU/ml<br />

were: 63%, 76%, 27% in naïve, relapsers and Nonresponders,<br />

respectively. At week 4 the PPV were:<br />

96% or 78%, 100% or 70%, 100 or 50% for TMA<br />

undetectable or 2 log drop, in naïve, relapsers and<br />

Non-responders, respectively.<br />

At week 12 the NPV were: 86% or 97%, 64% or 75%, 91<br />

or 93% for TMA undetectable or 2 log drop, in naïve,<br />

relapsers and Non-responders, respectively. Factors<br />

significantly associated with RVR odds ratio (95%CI)<br />

were genotypes 2-3: 6.5(6.3.4-12.2)(p


RESULTS: Youden indices derived from ROC curves<br />

increased from 0.26–0.34 at Week 4 to 0.41–0.55 at<br />

Week 32 in HBeAg-positive patients. Associated HBV<br />

DNA threshold values decreased from 5.1–5.6 log 10<br />

copies/mL at Week 4 to 2.7–3.1 logs at Week 24. In<br />

HBeAg-negative patients, at Week 24 Youden indices<br />

were maximized (0.34–0.49) and HBV DNA threshold<br />

values were minimized (2.5 log 10<br />

copies/mL).<br />

At Weeks 8 and 12, the predictive sensitivity of HBV<br />

DNA level is low, and specificity is high. At Weeks 24<br />

and 32, sensitivity is substantially higher, consistent<br />

with the marked increase in PCR-negativity at those<br />

points. NPV increased from Weeks 4 through 32 for all<br />

outcomes. Although the sensitivity of PCR-negativity<br />

for predicting no resistance increased from 0.63 to<br />

0.69 (HBeAg-positive) or from 0.87 to 0.97 (HBeAgnegative)<br />

from Weeks 24–32, specificity decreased<br />

from 0.78 to 0.76 (HBeAg-positive), or from 0.62<br />

to 0.46 (HBeAg-negative), indicating a decreased<br />

ability to assess resistance risk. Multivariate analyses<br />

of baseline HBV DNA, ALT, BMI, age, Ishak fibrosis<br />

score, Knodell HAI score, gender, and HBV genotype<br />

indicated that baseline HBV DNA level was the<br />

strongest predictor of virologic outcome at Week 24<br />

in both HBeAg-positive and negative patients.<br />

CONCLUSIONS: Week 24 is the best time point<br />

to assess initial response to telbivudine therapy,<br />

as it provides a favorable balance of sensitivity and<br />

specificity for identifying patients at risk of negative<br />

outcomes at 2 years. Thus clinical decisions to either<br />

continue or modify therapy can be made as early as<br />

Week 24.<br />

Abstract 13<br />

Green Fluorescence Based Assays<br />

for Hepatitis C Virus Replication<br />

and Infectivity in Cell Culture<br />

S Hazari, RF Garry, T Wakita, and S Dash<br />

Department of Pathology and Lab Medicine, Microbiology<br />

and Immunology, Tulane University Health Sciences<br />

Center, New Orleans, LA 70112, USA<br />

Background: A convenient and reliable assay<br />

system is needed for the measurement of hepatitis C<br />

virus replication and viral infectivity in a cell culture.<br />

Recently, a unique HCV clone derived from Japanese<br />

patients has been demonstrated to replicate efficiently<br />

in cell culture and produce infectious HCV particles.<br />

Aim: We sought to develop chimeric clones between<br />

green fluorescence protein (GFP) with full-length and<br />

sub-genomic JFH-1 clones such that HCV replication<br />

and infection can be examined directly using<br />

fluorescence microscopy.<br />

Methods: Chimeric full-length (JFH-1) and replicon<br />

(pSGR-JFH-1) clones were prepared by inserting<br />

the GFP coding sequences in frame with the coding<br />

sequences of HCV NS5A protein of JFH-1 clone.<br />

HCV RNA transcripts were prepared from each clone<br />

by T7 RNA polymerase and transfected to Huh-7<br />

cells by the electroporation method. The success<br />

of HCV transfection and replication was examined<br />

by developing stable replicon cell lines and in vitro<br />

infectivity of fluorescence virus particles using Huh-<br />

7.5 cells.<br />

Results: Both the replicon and full-length chimeric<br />

clone showed a fairly high-level expression of NS5A-<br />

GFP fusion protein that visualized by fluorescence<br />

microscopy and fusion protein can be detected by<br />

Western blot analysis. The replication of chimeric<br />

clones was confirmed by detecting positive strand<br />

HCV RNA by ribonuclease protein assay and the<br />

ability to form G-418 cell colonies. We now have<br />

14 Global Antiviral Journal Volume 3, Supplement 2


established several stable cell lines that support<br />

high-level replication of GFP-tagged sub-genomic<br />

HCV RNA. The level of viral replication between the<br />

wild type and chimeric clone is comparable. Huh-7<br />

cells transfected with GFP tagged viral genomes<br />

produced infectious virus particles since expression<br />

of GFP was demonstrated in naïve Huh-7.5 cells after<br />

virus infection. As a practical validation, we showed<br />

that replication and infectivity totally abolished by<br />

treatment with alpha interferon.<br />

Conclusion: We established a HCV cell culture<br />

systems using GFP tagged viral genomic and subgenomic<br />

clones that allow direct visualization of<br />

infected cells by fluorescence microscopy. These<br />

systems provide a powerful tool for the understanding<br />

of host-virus interactions and may provide a<br />

reliable model system to test varieties of anti-HCV<br />

substances.<br />

Abstract 14<br />

Development of Mouse Model for<br />

Hepatitis C Virus Replication<br />

S Dash, S Hazari, K Zackson, RF Garry, M Burrow, and<br />

T Mandal<br />

Department of Pathology and Laboratory Medicine,<br />

Microbiology, Medicine, Tulane University Health<br />

Sciences Center, Pharmacy Department, Xavier University<br />

of Louisiana, New Orleans, LA, USA<br />

Background: We previously published that<br />

intracellular immunization with recombinant<br />

antibodies to viral helicase and small interfering RNA<br />

can effectively eliminate hepatitis C virus replication<br />

and expression in vitro cell culture models. We realized<br />

that a reliable small animal model is needed to test<br />

the success of the intracellular immunization strategy<br />

against hepatitis C virus in vivo.<br />

Methods: We prepared interferon sensitive Huh-7<br />

cell line replicating green fluorescence (GFP)-tagged<br />

HCV sub-genomic RNA. Mouse adapted GFPtagged<br />

HCV replicon cell clone was isolated from<br />

subcutaneous tumors and serially passaged in SCID/<br />

bg mice followed by selection with G-418. Replicon<br />

cells (S-3/GFP) were implanted subcutaneously into<br />

gamma irradiated SCID mice to form a subcutaneous<br />

tumor. This model was validated by subcutaneous<br />

injection of interferon alpha (15,000 IU) daily up to<br />

2 weeks.<br />

Results: Highly tumorgenic sub clone of<br />

original interferon sensitive replicon (S-3/GFP)<br />

was isolated that express high levels of GFP after<br />

four passages in SCID mice. These replicon cells<br />

developed subcutaneous tumors in gamma irradiated<br />

SCID/bg mice in approximately two weeks. High<br />

levels of HCV replication in the Xenograft tumors<br />

were confirmed by detection of HCV positive<br />

strand RNA by ribonuclease protection assay (RPA)<br />

and GFP expression by fluorescence microscopy.<br />

Interferon alpha treatment completely cleared HCV<br />

RNA replication and expression in the subcutaneous<br />

tumors within two weeks.<br />

Conclusions: We prepared a mouse adapted GFPlabeled<br />

replicon cell line that formed a subcutaneous<br />

tumor and supported high levels of HCV replication<br />

in SCID mice. HCV replication in the subcutaneous<br />

tumors can be effectively inhibited by interferon<br />

alpha suggesting that this mouse model can be used<br />

to test novel therapeutics against HCV.<br />

Aim: To develop a mouse model in which HCV<br />

replication can be studied for a short-term in<br />

Xenograft tumor.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 15


Abstract 15<br />

A Cell-Based, Miniaturized<br />

Infection System to Identify<br />

Bioactive Molecules Against<br />

Hepatitis C Virus<br />

P Gastaminza 1 , A Montero 2 , S Pitram 2 , R Ghadiri 2 ,<br />

V Fokin 2 , B Sharpless 2 , and FV Chisari 1<br />

1 Department of Molecular and Experimental Medicine;<br />

2 Department of Chemistry, The Scripps Research<br />

Institute, 10550 N. Torrey Pines, La Jolla, CA 92037, USA<br />

BACKGROUND: In the past, the lack of a cell culture<br />

and small animal infection models has limited the<br />

discovery of new drugs in the field. Although the<br />

establishment of surrogate models (e.g. replicons,<br />

pseudoparticles) has greatly contributed to the<br />

discovery of potential new drugs, only limited<br />

aspects of the virus replication cycle can be targeted<br />

with those systems. The recent development of a<br />

robust model of HCV infection in cell culture allowed<br />

rescuing a recombinant genotype 2a virus from a<br />

consensus cDNA cloned from a japanese patient with<br />

fulminant hepatitis (JFH-1) and reproduction of the<br />

entire replication cycle of HCV in vitro.<br />

METHODS: Taking advantage of this new system<br />

we designed a simple yet sensitive and efficient<br />

colorimetric screening assay that allows evaluation<br />

of viral spread in a 96-well format. This technology<br />

permits the screening of large libraries to identify<br />

compounds that can target any aspect of the viral<br />

replication cycle, thus greatly increasing the potential<br />

yield of candidate antiviral molecules for further<br />

development.<br />

RESULTS: Applying this methodology, the screening<br />

of various chemical libraries including peptides and<br />

small molecules allowed the discovery of several<br />

new classes of molecules with antiviral activity in<br />

the submicromolar to low-micromolar range. The<br />

significance of the various hits obtained in the<br />

primary screening was evaluated by determining<br />

the potency and toxicity of the candidate molecules.<br />

Time-of-addition experiments as well as specific<br />

studies performed in surrogate models of HCV<br />

infection completed the characterization of the<br />

identified molecules and provided insights on the<br />

mode of action of the new antiviral compounds.<br />

CONCLUSION: We have developed a miniaturized<br />

HCV infection assay that allows screening chemical<br />

libraries for compounds with antiviral capacity.<br />

By reproducing the entire HCV replication cycle<br />

in vitro, this assay creates the unique opportunity<br />

to identify novel cellular and viral targets and<br />

therefore novel inhibitors of HCV infection since<br />

aspects of the viral replication cycle that could not<br />

be examined in previous screening assays (e.g. viral<br />

assembly, trafficking and secretion) can be efficiently<br />

interrogated in this system. Using this technology a<br />

new class of inhibitory peptides and three classes of<br />

novel small molecules where shown to significantly<br />

inhibit HCV infection.<br />

Abstract 16<br />

Generation of HCV E2 Specific<br />

Neutralizing Monoclonal Antibody<br />

SJ Park 1 , YJ Lee 1 , ET Park 1 , SH Lee 1 , SY Seol 1 ,<br />

HJ Won 2 , YJ Jeong 2 , SG Park 2 , IH Choi 2 , JW Shin 3 ,<br />

and NH Park 3<br />

1 Department of Internal Medicine; 2 Department of<br />

Microbiology, College of Medicine, Inje University, Busan;<br />

3 Department of Internal Medicine, University of Ulsan<br />

College of Medicine, Ulsan University Hospital, Ulsan,<br />

Korea<br />

Objectives: This study aimed to develop a human<br />

monoclonal antibody neutralizing hepatitis C virus<br />

infection from naïve human antibody library with<br />

phage display techniques.<br />

Methods: For selection of antibody clones reacted<br />

to HCV E2 protein, panning and screening was<br />

carried out with E2 protein immobilized CM5 chip in<br />

BIAcore 2000 from naïve antibody library sized 1 x<br />

16 Global Antiviral Journal Volume 3, Supplement 2


10 10 cfu. Selected antibody clones were analyzed by<br />

nucleotides sequencing and affinity measurements.<br />

Neutralizing activity was determined with HCVpp<br />

infection test into Huh7 cells.<br />

Results: Four clones (A4, F4, G4, and H6) were<br />

selected after panning and screening. A4 clones<br />

showed the highest affinity (9.60 x 10 -8 M -1 ). In the<br />

neutralizing assay, 10 µg/ml of A4 scFv was inhibited<br />

30% of HCVpp infection.<br />

Conclusions: This study generates human<br />

monoclonal antibody fragment, A4, neutralizing<br />

HCV E2 proteins. It might be useful in the passive<br />

immunotherapy for hepatitis C.<br />

Abstract 17<br />

A Mutation within the Hepatitis<br />

C Virus NS3 Helicase Domain<br />

Promotes Virion Assembly in<br />

Cultured Hepatoma Cells<br />

M Yi, Y Ma, J Yates, and SM Lemon<br />

The Center for Hepatitis Research, Institute for<br />

Human Infections and Immunity and Department of<br />

Microbiology and Immunology, University of Texas<br />

Medical Branch, Galveston, TX, USA<br />

BACKGROUND: A unique substitution in the amino<br />

acid sequence of the hepatitis C virus (HCV) NS3<br />

protein (Q1251L, Yi et al., J. Virol 81:629-38, 2007)<br />

rescues a defect in production of infectious virus in<br />

cells transfected with a chimeric RNA (H-NS2/NS3-J,<br />

designated “HJ3”) comprising the genotype 1a core-<br />

NS2 sequence placed within the background of the<br />

genotype 2a JFH-1 sequence. This is surprising as<br />

NS3, which possesses protease, helicase and NTPase<br />

activities, has not been suggested previously to play<br />

a role in virus assembly or release. We have studied<br />

how the Q1251L mutation contributes to infectious<br />

virus production by this chimera in order to better<br />

understand the role of NS3 in this process.<br />

METHODS: We assessed the effect of the<br />

Q1251L mutation on RNA replication by two<br />

different methods; quantitative Taqman RT-PCR<br />

measurements of genomic RNA, and a reporter assay<br />

using subgenomic replicon constructs. We measured<br />

the titer of intracellular and extracellular infectious<br />

virus produced from HJ3 RNA with or without the QL<br />

mutation to determine whether this mutation affects<br />

virus assembly and/or release. Similar experiments<br />

were performed using JFH1 and a second H77-JFH1<br />

chimera (H-(NS2)-J, designated HJ2). To further<br />

understand the NS3-mediated assembly process,<br />

we analyzed cell lysates by centrifugation through<br />

equilibrium density gradients and rate-zonal velocity<br />

gradients, determining the quantity of core protein<br />

and infectious virus in each fraction.<br />

RESULTS: Although Q1251 is highly conserved<br />

across HCV genotypes, the Q1251L substitution<br />

had no effect on replication of subgenomic replicon<br />

RNA, and did not impair NS3-associated enzymatic<br />

activities including processing at the NS2-NS3<br />

junction. However, it profoundly influenced assembly<br />

of the chimeric virus. While transfection of unmodified<br />

HJ3 RNA failed to produce either extracellular or<br />

intracellular infectious virus, transfection of HJ3<br />

RNA containing the Q1251L substitution (HJ3/QL)<br />

resulted in rapid and efficient release of infectious<br />

virus, which was also detected in cell lysates.<br />

Quantitative assessment of core protein abundance<br />

in fractions of isopycnic gradients loaded with lysates<br />

of transfected cells confirmed that core expressed<br />

by HJ3 did not assemble into rapidly sedimenting<br />

particles in the absence of the Q1251L substitution,<br />

suggesting a role for NS3 early in particle assembly.<br />

While the introduction of the Q1251L substitution<br />

into the parental JFH-1 sequence enhanced<br />

production of JFH-1 virus, it had no significant effect<br />

on assembly or release of a second chimeric virus,<br />

H/J2. This differs from H/J3 only in the C-terminal<br />

19 amino acid residues of NS2, providing genetic<br />

evidence for an interaction between NS3 and NS2<br />

during virus assembly.<br />

CONCLUSION: These results indicate a previously<br />

unsuspected role for NS3 in early virion morphogenesis<br />

and provide a new perspective on assembly of the<br />

infectious HCV particle.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 17


Diagnosis and New Treatments<br />

for HBV and HCV<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 19


Abstract 18<br />

HCV Replicons vs. Infectious Virus<br />

Systems in Drug Discovery<br />

SM Lemon<br />

Center for Hepatitis Research, University of Texas Medical<br />

Branch, Galveston, TX 77551-1073, USA<br />

The development of subgenomic and subsequently<br />

genome-length HCV replicons provided highly<br />

needed tools for antiviral drug discovery efforts.<br />

These autonomously replicating RNAs allowed<br />

early validation of the antiviral activity of NS3/4A<br />

protease and NS5B polymerase inhibitors that were<br />

active in cell-free enzyme assays, and in screening<br />

assays have led to the identification of compounds<br />

that hit novel targets within the HCV replicase, such<br />

as NS5A, for which there are no cell-free, enzymatic<br />

assays. However, while replicon models recapitulate<br />

most if not all aspects of HCV RNA replication,<br />

they are typically di-cistronic and place translation<br />

of the nonstructural proteins under control of a<br />

heterologous internal ribosome entry site. Moreover,<br />

they are selected for amplification capacity and<br />

typically contain one or more cell culture-adaptive<br />

mutations that generally limit replication of virus<br />

in the chimpanzee. While such mutations may have<br />

relatively little relevance for protease and polymerase<br />

inhibitors, their presence is a concern in evaluating<br />

compounds that target other nonstructural proteins<br />

with less well-defined roles in the virus replication<br />

cycle. In addition, replicons are of no use in efforts to<br />

target virus attachment and entry, or later steps in the<br />

assembly and egress of virus from infected cells. These<br />

steps in the virus replication cycle can be targeted<br />

using more recently available cell culture-infectious<br />

virus systems, but not without significant limitations.<br />

The genotype 2a JFH-1 virus has a robust replication<br />

phenotype, and viable chimeric viruses have now<br />

been constructed that express the structural proteins<br />

of the more prevalent and relatively interferonresistant<br />

genotype 1a and 1b viruses from within<br />

the JFH-1 background. These new viruses represent<br />

valuable tools for the characterization of virus entry<br />

and the development of entry inhibitors, yet the<br />

buoyant density of these infectious virus particles<br />

is significantly less than that of virus produced<br />

within the liver in situ, indicating a need for caution<br />

in extrapolating from results obtained with cell<br />

culture-derived virus. The genotype 1a H77S virus<br />

has a substantially less robust replication phenotype<br />

than JFH-1 and its related inter-genotypic chimeras.<br />

However, in its most recent stage of development,<br />

cells transfected with H77S.2 RNA release ~10 3 FFU/<br />

ml of infectious virus into supernatant fluids, and<br />

this virus is capable of limited serial cell-free passage.<br />

While not suitable for high throughput screening,<br />

H77S.2 is useful in validation experiments as it is<br />

comprised of entirely genotype 1a sequence with<br />

defined cell-culture adaptive mutations.<br />

Abstract 19<br />

The Size of the Viral Inoculum<br />

Determines the Kinetics,<br />

Magnitude and Outcome of<br />

Hepatitis B Virus Infection<br />

S Asabe 1 , SF Wieland 1 , R Engle 2 , RH Purcell 2 , and<br />

FV Chisari 1<br />

1 Scripps Research Institute, La Jolla, CA, USA;<br />

2 National Institutes of Health, Bethesda, MD, USA<br />

The factors that determine the outcome of hepatitis<br />

B virus (HBV) infection include the age of onset of<br />

the infection and the immune responsiveness of the<br />

host. Little is known about the impact of virological<br />

determinants in this regard. Based on precedent in<br />

other viral systems, it is assumed that high antigen<br />

burden will exhaust or otherwise suppress the<br />

immune response and, thus, favor viral persistence.<br />

To test that hypothesis we examined the impact of<br />

inoculum size on the outcome of HBV infection<br />

in experimentally infected chimpanzees. Animals<br />

were injected intravenously with 10 10 , 10 7 , 10 4 , 10 1<br />

or 10 0 genome equivalents (GE) of a monoclonal<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 21


HBV inoculum and monitored for 1 year thereafter.<br />

Animals receiving 10 10 , 10 7 , and 10 4 GE of HBV<br />

developed self-limited infections characterized by<br />

dose-related onset kinetics and magnitude of infection<br />

that resolved within 12-16 weeks of inoculation.<br />

Resolution of infection in these animals was preceded<br />

by an early HBcAg-specific CD4 T cell response and<br />

coincided with a highly synchronized, early interferon<br />

gamma-producing T cell response and sharply elevated<br />

serum alanine aminotransferase (ALT) activity.<br />

Unexpectedly, virus spread to 100% of the hepatocytes<br />

in all animals that were inoculated with 10 1 or 10 0<br />

GE of HBV, and they developed either chronic (>52<br />

weeks) infections with the histological characteristics<br />

of chronic active hepatitis, or greatly prolonged (36-<br />

42 weeks) infections that ultimately resolved in the<br />

setting of an ALT flare. The immunological correlates<br />

of these unexpected outcomes were the absence of<br />

early CD4 T cell responses to HBcAg, the delayed<br />

onset of poorly synchronized, interferon gammanonproducing<br />

peripheral CD8 T cell responses, and<br />

the induction of delayed but vigorous interferon<br />

gamma-producing intrahepatic T cell responses that<br />

were functionally unable to terminate the infection.<br />

Interestingly, serum IL10 and intrahepatic PD1 mRNA<br />

levels were higher and sustained in the persistent/<br />

prolonged low dose infections than in the rapidly<br />

controlled high dose infections. These results suggest<br />

that subviral antigens in the infectious inoculum may<br />

prime antiviral T cell responses that precede viral<br />

spread through the liver and prepares the host for an<br />

anamnestic response that controls the infection when<br />

viral antigens are subsequently produced by infected<br />

cells. In contrast, failure to prime the T cell response<br />

early in HBV infection results in uncontrolled viral<br />

spread in the liver where intrahepatic T cell priming<br />

and persistent high level antigen production induce<br />

negative immunological regulators (e.g. IL10 and<br />

PD1) that further impair CD8 T cell function and lead<br />

to prolonged or persistent infection.<br />

Abstract 20<br />

Population Genetics of HBV and<br />

HCV Quasispecies: Implications<br />

for Drug Development and<br />

Drug-Resistance Testing<br />

RW Shafer<br />

Departments of Medicine and Pathology, Stanford<br />

University, Stanford, CA, USA<br />

BACKGROUND: HBV and HCV, like HIV, share the<br />

following characteristics: (i) they each cause a chronic<br />

life-threatening disease, (ii) they are each treatable<br />

with small molecular weight compounds, and (iii)<br />

they each have highly error-prone mechanisms<br />

of replication causing them to exist in vivo as a<br />

quasispecies – or mixture of innumerable related<br />

sequence variants.<br />

METHODS: My laboratory has been using ultra-deep<br />

pyrosequencing (UDPS; 454 Life Sciences, Bradford<br />

CT), to characterize the extent of genetic variability in<br />

the molecular targets of HIV, HBV, and HCV therapy<br />

in treatment-naïve and treatment-experienced<br />

individuals. My presentation will focus on our labs<br />

results using UDPS for sequencing HBV and HIV and<br />

on our plans for using UDPS for sequencing HCV. I<br />

will also review the main UDPS experimental design<br />

choices including (i) approaches to maximize the<br />

number of sequencable virus templates, (ii) the choice<br />

of sequencing strategy: “shotgun” vs tailed primers,<br />

(iii) the relative importance of PCR enzyme fidelity,<br />

and (iv) statistical analyses for handling random<br />

errors and G to A hypermutation.<br />

RESULTS: We have sequenced the RT and protease<br />

quasispecies in 48 clinical plasma HIV-1 isolates and<br />

a 1.2 bp pol region in 32 clinical plasma HBV isolates.<br />

Our first 32 HIV-1 isolates were sequenced using the<br />

first generation, GS20 sequencing platform. Each of<br />

the HBV isolates and our most recent HIV-1 isolates<br />

were sequenced using he second generation, FLX<br />

sequencing platform. In each experiment, we have<br />

22 Global Antiviral Journal Volume 3, Supplement 2


characterized the UDPS error rate using plasmid<br />

clones and have developed a robust statistical<br />

approach to distinguishing authentic minor<br />

variants from possible sequencing errors. For most<br />

experiments, the sensitivity for detecting minor<br />

variants which ranged from 0.2% to 2.0%, was<br />

primarily limited by the number of templates that<br />

could be introduced into the sequencing reaction<br />

(rather than by the sequencing technology itself). HIV<br />

and HBV infected persons receiving antiviral therapy,<br />

were consistently found by UDPS to have clinically<br />

relevant drug-resistant mutations detected at levels<br />

ranging from 1% to 20% that were rarely detected<br />

by standard population-based sequencing. Extensive<br />

molecular and limiting dilution clonal sequencing,<br />

furthermore, confirmed that the vast majority, if not<br />

all of these minor variants, were authentic.<br />

CONCLUSIONS: In theory, UDPS is an eminently<br />

logical approach to characterizing genetic variability<br />

and detecting drug-resistance mutations at the<br />

earliest stages of their development. In practice,<br />

however, both the upstream processing of samples<br />

prior to UDPS and the downstream analysis of UDPS<br />

sequence data are uncharted areas that require<br />

considerable amounts of optimization before this<br />

technology can achieve the reliability that currently<br />

exists for standard dideoxynucleoside sequencing.<br />

Abstract 21<br />

Nitazoxanide: A Potent Antiviral<br />

Agent Against HCV and HBV<br />

BE Korba 1 , AB Montero 1 , JS Glenn 2 , MS Ayers 3 , and<br />

J-F Rossignol 3<br />

1 Dept. of Microbiology & Immunology, Georgetown<br />

Univ. Med. Ctr., Washington, DC, USA; 2 Division of<br />

Gastroenterology & Hepatology, Dept. of Med., Stanford<br />

Univ. Sch. of Med., Stanford, CA, USA; 3 The Romark<br />

Institute for Medical Research, Tampa, FL , USA<br />

Nitazoxanide (NTZ), a thiazolide, is a broad spectrum<br />

anti-infective drug marketed in the United States<br />

(Alinia ® ) for treating gastroenteritis caused by<br />

Cryptosporidium parvum and Giardia lamblia, and is in<br />

clinical development for treating Clostridium difficile<br />

and rotavirus-associated diseases. NTZ and its active<br />

circulating metabolite, tizoxanide (TIZ), exhibited<br />

potent and selective antiviral effects in cell culture<br />

models of HCV and HBV replication (Korba, et al.,<br />

2007, Antivir. Res., in press). Both NTZ and TIZ were<br />

effective against HCV replication in both genotype<br />

1b (CON1) and 1a (H77) replicon cell lines. Moderate<br />

synergistic interactions against HCV were observed<br />

between NTZ and either interferon alpha 2b (IFN), or<br />

2’C-methyl cytidine in combination treatments. NTZ<br />

was equally effective against NS5B S282T, and NS3<br />

A156S/V/T drug-resistant mutants. Pretreatment of<br />

cultures with NTZ monotherapy further enhanced<br />

the potency of NTZ + IFN (but not NTZ + 2’CmeC)<br />

combinations. Although it was possible to select<br />

NTZ and TIZ resistant HCV replicon cell lines, the<br />

resistance phenotype was not transferred to naïve<br />

cells by transfection of HCV RNA from these cell<br />

lines. Both compounds also inhibited intracellular<br />

HBV replication and virion production in 2.2.15<br />

cells. NTZ exhibited moderately synergistic anti-<br />

HBV interactions with either of two licensed anti-<br />

HBV agents, lamivudine (LMV) or adefovir dipovoxil<br />

(ADV), and was equally effective in inhibiting the<br />

replication of several LMV and ADV-resistant<br />

mutants. Most notably, NTZ induced a reduction<br />

in the production of several HBV proteins (HBsAg,<br />

HBeAg, HBcAg) without a corresponding reduction<br />

in HBV RNA, indicating a post-transcriptional/<br />

translation mechanism.<br />

Clinical trials against chronic HCV infection are<br />

in progress in the USA and Egypt. Interim reports<br />

on clinical efficacy (STEALTH C-1 trial) have been<br />

recently presented (Rossignol, et al., 2007, Hepatol.<br />

46(Suppl. 1):316A). In naïve patients, combination<br />

therapy consisting of 12 weeks pretreatment with<br />

NTZ followed by 36 weeks of NTZ in combination<br />

with PegIFNα-2a+ribavirin was significantly more<br />

effective than 48 weeks of PegIFNα-2a+ribavirin<br />

(standard of care) (SVR12, 79% vs. 45%, p=0.007).<br />

Detailed studies of mechanisms against both HBV and<br />

HCV are currently under investigation. Preliminary<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 23


data indicate that host cell processes are targeted,<br />

most likely components of the unfolded protein<br />

and/or ER stress response pathways. Current data<br />

indicate that the intracellular environment induced<br />

by each virus may influence the consequences of druginduced<br />

changes in cellular responses. Nitazoxanide<br />

is a promising new antiviral agent that, due to its<br />

probable novel mechanism of action, has substantial<br />

potential for use as an adjunct to current and future<br />

therapies to enhance sustained response rates against<br />

chronic hepatitis virus infection and disease.<br />

Abstract 22<br />

Clinical Implications of Combined<br />

Intra-Cellular and Cellular<br />

Evolution of HCV Resistance<br />

during Direct Anti-HCV<br />

Treatment<br />

AU Neumann and J Guedj<br />

Bar-Ilan University, Ramat-Gan, Israel<br />

Background and goal: The current paradigm for<br />

modeling development of viral resistance, based on<br />

the HIV experience, considers evolution of resistance<br />

only on the cellular infection level. While this is correct<br />

for HIV, a retrovirus RNA virus for which mutation<br />

occurs mainly at the RT step, it is known that for<br />

HCV all processes of resistance evolution – mutation,<br />

selection and amplification – can occur on a faster<br />

time-scale of RNA synthesis at the intra-cellular level.<br />

Here we explore, with a novel mathematical model<br />

that considers both intra-cellular level evolution and<br />

cellular infection level viral dynamics, the clinical<br />

implication of intra-cellular resistance evolution for<br />

direct anti-HCV drugs.<br />

Methods: In the model, intra-cellular RNA (ICR)<br />

is used to form replication units (RU), which in turn<br />

synthesize more ICR that is partially packaged and<br />

secreted as virions. Direct anti-HCV drugs can have<br />

an anti-viral effect through blocking of RU formation,<br />

ICR synthesis and/or virion export. The development<br />

of resistance is modeled in the intra-cellular level by<br />

the evolution and dynamics of different strains of RU<br />

and ICR with different drug-sensitivity and different<br />

relative-fitness. Resistance evolution also impacts the<br />

cellular infection level as result of the exported virus<br />

of different strains.<br />

Results: Evolution of resistant virus is faster when<br />

it occurs at the intra-cellular level as compared to<br />

occurring only at the cellular infection level, assuming<br />

similar rates of mutation, and similar distribution of<br />

sensitivity to drug and relative fitness of the different<br />

strains in both models. An analytical estimate - based<br />

on the relative-fitness, drug sensitivity and mutation<br />

rate parameters - was obtained for the time it takes<br />

a resistant strain to reach equal concentration as the<br />

wild-type, and for the resistance rebound slope.<br />

The combination of resistance evolution on the intracellular<br />

level with cellular infection viral dynamics<br />

level allows for more complex viral kinetic patterns<br />

than possible with resistance evolution on the cellular<br />

infection level only. In general, a complete rebound, a<br />

tri-phasic decline, a bi-phasic decline with a shoulder,<br />

a decline at the delta mode (with the rate of infected<br />

cell loss) or a decline at the gamma mode (with the<br />

faster rate of intra-cellular RU loss) are all possible<br />

as function of the mutation rate and the distribution<br />

of fitness and sensitivity. Of particular interest, we<br />

predict that it is possible for the total viral load to<br />

decline (in the delta mode, after a transient rebound)<br />

even after takeover by a virus strain fully resistant to<br />

a drug given as a mono-therapy.<br />

Conclusions: More rapid and more complex<br />

patterns of viral resistance evolution are predicted<br />

when considering HCV resistance evolution at the<br />

intra-cellular level together with cellular level viral<br />

dynamics. Some of the patterns predicted by the<br />

model were already observed in data publicly released<br />

for different direct anti-HCV drugs. The model<br />

makes several predictions with important clinical<br />

implications, such as the possibility for decline<br />

with fully resistant virus during monotherapy, and<br />

analytical estimates allow long-term prediction based<br />

on early kinetic information.<br />

24 Global Antiviral Journal Volume 3, Supplement 2


Abstract 23<br />

Phase 2 Studies with<br />

Albinterferon alfa-2b (alb-IFN)<br />

Dosed q4wk Provide Insights into<br />

Dose Selection for Future Studies<br />

I Jacobson 1 and D Nelson 2<br />

1 Weill Medical College of Cornell University, New York,<br />

USA; 2 University of Florida at Gainesville, Florida, USA<br />

BACKGROUND: alb-IFN is a novel, long-acting<br />

interferon (IFN) with a half-life of ~150 h, achieving<br />

substantial drug exposure levels, and the capability<br />

of maintaining antiviral activity detected over a 28-d<br />

dosing interval. Phase 2 studies have been conducted<br />

in IFN-naïve patients with chronic hepatitis C (CHC)<br />

to assess the efficacy and safety of q4wk dosing.<br />

METHODS: Study 1 (genotype [Gt] 1, CHC): alb-IFN<br />

1200 μg q4wk (n = 116) was compared with alb-IFN<br />

900 and 1200 μg q2wk (n = 118 and 110, respectively),<br />

and peginterferon alfa-2a (PEG-IFNα-2a) 180 μg<br />

qwk (n = 114), plus ribavirin 1000-1200 mg/d in all<br />

arms. Study 2 (Gt 2/3, CHC): alb-IFN 1500 μg q4wk<br />

(n = 22) was compared with alb-IFN 1500 μg q2wk<br />

(n = 21), plus ribavirin 800 mg/d. Primary efficacy<br />

endpoint was sustained virologic response (SVR) in<br />

both studies.<br />

RESULTS: In study 1, the SVR rates with alb-IFN<br />

1200 μg q4wk by intent-to-treat analysis vs alb-IFN<br />

900 μg q2wk and PEG-IFNα-2a were 51% vs 58% and<br />

58%, respectively (P = .28 vs PEG-IFNα-2a). Although<br />

rapid virologic response rate at wk 4 (hepatitis C virus<br />

[HCV] RNA < limit of quantitation [43 IU/mL]) was<br />

lower in patients with alb-IFN 1200 μg q4wk vs alb-<br />

IFN 900 μg q2wk and PEG-IFNα-2a (18% vs 25%<br />

and 26%, respectively), those patients had a 100%<br />

positive predictive value of achieving SVR. Viral<br />

breakthrough and relapse rates with alb-IFN 1200 μg<br />

q4wk were comparable to the q2wk and PEG-IFNα-2a<br />

arms. alb-IFN 1200 μg q4wk was associated with less<br />

hematologic toxicity, which was reflected in the need<br />

for fewer dose reductions. In study 2, alb-IFN 1500<br />

μg q4wk resulted in an SVR rate of 77% vs 62% for the<br />

q2wk regimen. Early response rates (HCV RNA


of at least 4 distinct non-nucleoside inhibitor (NNI)-<br />

binding sites on the HCV polymerase. We previously<br />

reported a series of indole-based HCV polymerase<br />

NNIs that bind the enzyme at an allosteric site<br />

found at the junction of the thumb domain and the<br />

N-terminal finger loop (“finger-loop” NNIs). Although<br />

inhibitors of this class have shown to be highly active<br />

in biochemical and cell-culture assays, evidence of in<br />

vivo antiviral activity associated with this mechanism<br />

of action was still missing.<br />

Methods: A finger-loop NNI of HCV genotype (gt)<br />

1 and gt 3 NS5B polymerases, with an IC 50<br />

values of<br />

30 nM against the genotype 1b replicon, was dosed<br />

for 5 days at 10 mg/kg bid orally to one chimpanzee<br />

infected with gt 1a HCV, and at 2 and 10 mg/kg to a<br />

second chimpanzee infected with gt 1b HCV.<br />

Results: We observed a 1.1 and 3.8 log 10<br />

viral load<br />

reduction in the gt 1b HCV-infected animal at doses<br />

of 2 and 10 mg/kg, respectively. Viremia rebounded<br />

after treatment ended. In the gt 1a HCV-infected<br />

chimp, a dose of 10 mg/kg elicited a 1.4 log 10<br />

drop<br />

in viral load in 48 h. Cloning and sequencing of the<br />

NS5B polymerase gene from the viral isolates present<br />

in the two different chimps did not reveal any obvious<br />

genetic reason for the difference in antiviral response,<br />

and only small differences in susceptibility to this<br />

class of inhibitors were observed with the polymerase<br />

genes from the two viral isolates when assayed<br />

in the replicon system. Moreover, no resistance<br />

mutations or treatment related sequence changes<br />

were identified. When antiviral response (log 10<br />

drop<br />

in viremia) observed in the 3 dosing intervals of the<br />

two chimps was analyzed as a function of the ratio<br />

of trough plasma concentration and normalized for<br />

replicon EC 50<br />

, a very steep dose-response curve was<br />

obtained. Thus, small differences in exposure/potency<br />

produced large effects on viremia.<br />

Conclusions: This study demonstrated that<br />

finger-loop NNIs are efficacious at inhibiting viral<br />

replication in the HCV infected chimpanzee model<br />

and suggests that the chimpanzee model could prove<br />

useful for determining the PK/PD relationship for<br />

experimental compounds with novel mechanism of<br />

action.<br />

Abstract 25<br />

Preclinical Development of the<br />

Amphipathic DNA Polymer REP<br />

9AC for the Treatment of HBV<br />

Infection<br />

F Noordeen 1 , A Vaillant 2 , JM Juteau 2 , and A Jilbert 1,3<br />

1 School of Molecular and Biomedical Science, The<br />

University of Adelaide, Australia; 2 REPLICor Inc, Laval,<br />

Quebec, Canada; 3 Infectious Diseases Laboratories,<br />

Institute of Medical and Veterinary Science, Adelaide,<br />

Australia<br />

Background: Amphipathic DNA polymers (APs)<br />

are a novel class of nucleic acid medicine based<br />

on phosphorothioate modified DNA which have a<br />

broad spectrum activity against enveloped viruses.<br />

In type 1 fusion viruses, APs interact with conserved<br />

amphipathic alpha helical domains on surface<br />

glycoproteins blocking their entry or release activities.<br />

APs are also effective against non-type 1 enveloped<br />

viruses such as HCV.<br />

Methods: The antiviral efficacy of the lead AP<br />

candidate REP 9AC, a 40mer poly AC phosphorothioate<br />

oligonucleotide, against another non-type 1 enveloped<br />

virus, the human hepatitis B virus (HBV), was studied<br />

using the duck hepatitis B virus (DHBV) model both<br />

in vitro in primary duck hepatocytes (PDH) and in<br />

vivo in the Pekin duck.<br />

Results: REP 9AC demonstrated potent antiviral<br />

activity against DHBV infection of PDH suggesting<br />

that a large amphipathic domain related to those<br />

found in type 1 fusion glycoproteins is involved in<br />

the entry or release of DHBV. In vivo, REP 9AC was<br />

assessed for its ability to block virus entry and cellto-cell<br />

spread of DHBV infection. Fourteen-day-old<br />

ducks were infected with a defined dose of DHBV<br />

and treated intraperitoneally with REP 9AC (10mg/<br />

kg/day) or orally with the Bristol-Myers Squibb<br />

nucleoside analogue, Entecavir (ETV; 1 mg/kg/day)<br />

for 14 days. REP 9AC was well tolerated by the ducks<br />

with no detectable side effects. Biopsy liver tissue<br />

26 Global Antiviral Journal Volume 3, Supplement 2


collected on day 4 post-inoculation (pi) showed a<br />

small percentage of DHBV-infected hepatocytes in<br />

all REP 9AC and control ducks. However, by day 14<br />

pi 100% of ducks treated with REP 9AC showed no<br />

evidence of continuing DHBV infection in the liver<br />

using immunohistochemistry for DHBV surface<br />

antigen, or Southern blot detection of DHBV DNA.<br />

As expected from previous studies treatment of ducks<br />

with ETV inhibited DHBV replication and prevented<br />

the cell-to-cell spread of DHBV infection, while in all<br />

saline treated ducks DHBV infection spread to infect<br />

>95% of hepatocytes (Foster et al., J Virol 2005). In<br />

a subsequent in vivo dose response experiment, REP<br />

9AC was similarly effective at doses as low as 1mg/<br />

kg/day.<br />

Conclusions: AP chemistry (phosphorothioated<br />

DNA) has an established record of being well<br />

tolerated in human patients and has a liver ½ life in<br />

excess of one month in human patients, suggesting<br />

that REP 9AC could be an effective treatment for<br />

HBV infection either as mono or combination<br />

therapy, which can likely be given once a week. More<br />

importantly, the novel mode of action of APs strongly<br />

suggest that treatment with REP 9AC will not result<br />

in the development of antiviral drug resistance. The<br />

compounds may also address the still uncertain<br />

role of virus spread in maintenance of chronic HBV<br />

infections, as well as preventing the spread of antiviral<br />

drug resistant HBV mutants in treated patients.<br />

Abstract 26<br />

Inhibition of Hepatitis C<br />

Virus Replication by<br />

Octadecyloxypropyl -(S)-HPMPA<br />

in Genotype 1B and 2A Replicons<br />

KY Hostetler, DL Wyles, KA Kaihara, JR Beadle and<br />

RT Schooley<br />

Division of Infectious Diseases, University of California,<br />

San Diego, La Jolla, CA, USA<br />

BACKGROUND: Alkoxyalkyl esters of (S)-9-[3-<br />

hydroxy-2-(phosphonomethoxy)-propyl]-adenine<br />

((S)-HPMPA) show broad spectrum antiviral activity<br />

against double stranded DNA viruses as well as HIV-1<br />

and hepatitis B viruses, and are orally active in animal<br />

models of poxvirus, cytomegalovirus and hepatitis<br />

B (HBV). The NS5B RNA polymerase of hepatitis<br />

C virus (HCV) is required for viral replication and<br />

several inhibitors are under evaluation. Inhibitors<br />

of this polymerase would be particularly attractive<br />

since the active site is highly conserved among HCV<br />

genotypes. We synthesized octadecyloxypropyl-(S)-<br />

HPMPA (ODE-(S)-HPMPA) and tested it against 1B<br />

and 2A replicons.<br />

METHODS: Genotype 1B and 2A replicons<br />

containing the firefly luciferase gene were transfected<br />

into Huh 7.5.1 cells. The cells were incubated with or<br />

without (S)-HPMPA or ODE-(S)-HPMPA for 48 to 72<br />

hours. Viral replication was assessed by measuring<br />

luciferase activity and cytotoxicity was measured with<br />

MultiTox-Fluor. The dose response data was analyzed<br />

and the EC 50<br />

and CC 50<br />

values were obtained by fitting<br />

a sigmoidal dose-response curve to the luciferase data<br />

using Prism software (GraphPad, v4). The selectivity<br />

indexes were calculated (CC 50<br />

/EC 50<br />

).<br />

RESULTS: Unmodified (S)-HPMPA was inactive<br />

in both 1B and 2A luciferase replicon systems<br />

(EC 50<br />

>100 µM). However, ODE-(S)-HMPA exhibited<br />

substantial antiviral activity in both 1B and 2A<br />

replicons with EC 50<br />

values in the 0.5 to 1.5 µM range.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 27


Selectivity indexes ranged from 25 to 50. We studied<br />

the cellular penetration and conversion of 14 C-labeled<br />

(S)-HPMPA and ODE-(S)-HPMPA to HPMPA-diphosphate<br />

in HepG2 cells in vitro. The absence of antiviral<br />

activity observed with (S)-HPMPA appears to be due<br />

to minimal cellular uptake and conversion of (S)-<br />

HPMPA to HPMPA-diphosphate; ODE-(S)-HPMPA<br />

uptake and conversion to HPMPA-diphosphate is<br />

many-fold greater than that of (S)-HPMPA itself.<br />

The mechanism of inhibition of viral replication is<br />

not known at the present time.<br />

CONCLUSIONS: The octadecyloxypropyl prodrug<br />

of (S)-HPMPA is a potent and selective inhibitor<br />

of HCV genotype 1B and 2A replicons. This is<br />

surprising since (S)-HPMPA is an analog of<br />

deoxyadenosine monophosphate and is known to<br />

inhibit double stranded DNA viruses. Its structure<br />

differs substantially from other nucleoside analog<br />

lead compounds identified to date which tend to<br />

be analogs of ribonucleotides. ODE-(S)-HPMPA is<br />

worthy of further evaluation since it has already been<br />

shown to be effective orally in animal models of HBV<br />

and would be expected to have HCV activity in vivo.<br />

Abstract 27<br />

FKBP8 Plays a Crucial Role in the<br />

Replication of Hepatitis C Virus<br />

T Okamoto, K Moriishi, and Y Matsuura<br />

Department of Molecular Virology, Research Institute for<br />

Microbial Diseases, Osaka University, Osaka, Japan<br />

BACKGROUND: Hepatitis C virus (HCV) nonstructural<br />

protein 5A (NS5A) is a component of<br />

the viral replication complex and is well known to<br />

modulate the functions of several host proteins.<br />

Although NS5A could be a candidate as the drug<br />

target for HCV, host factors that associate with NS5A<br />

for HCV replication have not been clarified yet.<br />

METHODS: To gain further insight into the functional<br />

role of NS5A in HCV replication, we screened human<br />

libraries by a yeast two-hybrid system using NS5A<br />

as bait and identified FKBP8, a member of the<br />

FK506-binding protein family, as an NS5A-binding<br />

protein. We further examined the host proteins<br />

interact with FKBP8 by immunoprecipitation and<br />

LC-MS/MS analyses. We investigated the biochemical<br />

characteristics and the intracellular localization of<br />

NS5A and FKBP8 by immunoprecipitation, surface<br />

plasmon resonance analysis, and fluorescence and<br />

electron microscopy.<br />

RESULTS: The siRNA-mediated knockdown of FKBP8<br />

in HCV RNA replicon cells suppressed RNA replication,<br />

and this reduction was reversed by the expression of<br />

an siRNA-resistant FKBP8 mutant. Furthermore, the<br />

knockdown of FKBP8 could also impair production of<br />

infectious virus in HCV cell culture system. Analysis<br />

of the cellular proteins interacting with FKBP8 by<br />

LC-MS/MS revealed that Hsp90 is an FKBP8-binding<br />

protein and forms a complex with FKBP8 and NS5A.<br />

The complex was shown to be critical for HCV<br />

replication, as based on the finding that treatment<br />

of the HCV replicon cells with geldanamycin, an<br />

inhibitor of Hsp90, suppressed RNA replication in a<br />

dose-dependent manner. Surface plasmon resonance<br />

analysis revealed that the dissociation constant of the<br />

interaction between the purified FKBP8 and NS5A<br />

expressed in bacteria was 82 nM. Mutational analyses<br />

of FKBP8 and NS5A indicated that three sets of<br />

tetratricopeptide repeats in FKBP8 are responsible for<br />

interactions with NS5A and that a single amino acid<br />

residue of Val or Ile at 121 position of NS5A is critical<br />

for the specific interaction with FKBP8. Substitution<br />

of the Val 121 to Ala drastically impaired the replication<br />

of HCV replicon cells, and the drug-resistant replicon<br />

cells emerging after drug selection were shown to have<br />

reverted to the original arrangement by replacing<br />

Ala 121 with Val. Examination of individual fields of<br />

the replicon cells by both fluorescent microscopy<br />

and electron microscopy (the correlative fluorescent<br />

microscopy-electron microscopy technique) revealed<br />

that FKBP8 is partially co-localized with NS5A in the<br />

cytoplasmic structure known as the membranous<br />

web.<br />

CONCLUSION: These results indicate that NS5A<br />

directly binds to FKBP8 through the Val 121 and FKBP8<br />

28 Global Antiviral Journal Volume 3, Supplement 2


further recruits Hsp90 to the replication complex.<br />

Recently, geldanamycin was shown to have a drastic<br />

impairment of the replication of poliovirus without any<br />

emergence of escape mutants. Therefore, disruption<br />

of the specific interaction of NS5A with FKBP8 might<br />

be an ideal target for a novel therapeutic of chronic<br />

hepatitis C with a low frequency of emergence of<br />

drug-resistant breakthrough viruses.<br />

Abstract 28<br />

Initial Recommendations for HCV<br />

Drug Resistance Analysis:<br />

A Consensus Statement from the<br />

HCV Drug Resistance Advisory<br />

Group<br />

K Lin 1 , D Hazuda 2 , M Otto 3 , C Boucher 4 , C Schooley 5 ,<br />

J O’Rear 6 , D Kempf 7 , A Kwong 8 , and N Parkin 9 for the<br />

HCV Drug Resistance Advisory Group<br />

1 Novartis, Cambridge, MA, USA; 2 Merck & Co., West<br />

Point, PA, USA; 3 Pharmasset, Princeton, NJ, USA;<br />

4 Universitair Medical Center, Utrecht, Netherlands;<br />

5 University of California, San Diego, San Diego, CA, USA;<br />

6 FDA/CDER/OND/OAP/ Division of Antiviral Products,<br />

Silver Spring, MD, USA; 7 Abbott, Abbott Park, IL, USA;<br />

8 Vertex Pharmaceuticals, Inc., Cambridge, MA, USA;<br />

9 Monogram Biosciences, South San Francisco, CA, USA<br />

Background: As new anti-HCV drugs, especially<br />

protease and polymerase inhibitors, are evaluated in<br />

clinical trials, interest in assays for monitoring the<br />

development of resistance to these agents and for<br />

avoiding cross-resistance has increased. However,<br />

there is little guidance about the nature (phenotypic<br />

or genotypic) of the resistance assays, when to use<br />

them, or desired performance characteristics. The<br />

HCV Drug Resistance Advisory Group (DRAG) was<br />

formed in late 2006 to formulate recommendations<br />

for methods to measure resistance and how such<br />

methods should be integrated into clinical trials, and<br />

potentially following drug approval.<br />

Methods: Knowledge about mutations associated<br />

with resistance to investigational protease inhibitors<br />

(PIs), nucleoside polymerase inhibitors (NIs), and<br />

non-nucleoside polymerase inhibitors (NNIs)<br />

was gathered from the literature and conference<br />

presentations. Recommendations for the design and<br />

use of HCV resistance assays were based on diagnostic<br />

assay validation standards, previous experience in<br />

the HIV drug resistance field, and consultation with<br />

regulatory agencies, hepatologists and infectious<br />

disease experts.<br />

Results: Based on exposure of HCV genotype 1 to<br />

PIs both in vitro and in vivo, positions in NS3 associated<br />

with resistance include 36, 54, 155, 156, 168 and<br />

170. NIs (4’-Azido cytidine and 2’-C-Me nucleosides)<br />

select for mutations in NS5B at positions 96 and 282<br />

(respectively). NNIs belong to one of 4 classes, based<br />

on chemical structure and binding site, and select for a<br />

multitude of mutations in the thumb or palm regions<br />

of NS5B, including 201, 316, 411, 414, 419, 423,<br />

448, 451, 495, 496, and 499. Mutations conferring<br />

resistance to inhibitors may pre-exist in viruses from<br />

naïve patients as polymorphisms. Issues surrounding<br />

the desired performance characteristics, validation<br />

criteria, and implementation during clinical trials of<br />

sequence analysis and phenotypic assays have been<br />

discussed in the DRAG and draft recommendations<br />

have been formulated. Sequence analysis can be<br />

performed on a population or clonal basis, depending<br />

on the question being addressed (dominant mutations<br />

or minority species, pre-existing resistant variants,<br />

and linkage of multiple mutations). Amplification<br />

sensitivity targets of approximately 1,000 RNA<br />

copies/ml are desired, and sensitivity for detection<br />

of minority species must take into account the viral<br />

load and number of amplifiable genome templates in<br />

the plasma sample. Phenotypic susceptibility assays<br />

are crucial for the understanding of resistance and<br />

interpretation of genotypic data; prototype assays<br />

based on genotype 1 replicons have been described<br />

and are currently a popular approach, complemented<br />

by enzymatic assays using protein expressed in<br />

recombinant or cell-based systems.<br />

Conclusion: The recommendations made by the<br />

HCV DRAG are a starting point for future discussions<br />

about the role of resistance testing for HCV and for<br />

the establishment of standardized validation methods<br />

and criteria.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 29


Abstract 29<br />

Early Biochemical and Virological<br />

Response of Clevudine Therapy<br />

in Patients with HBV Associated<br />

Liver Cirrhosis<br />

KW Chung 1 , CY Ha 1 , SJ Baik 1 , CH Lee 2 , JS Lee 3 ,<br />

HJ Lee 4 , BH Han 5 , WC Choi 6 , SW Paik 7 , KC Koh 7 ,<br />

JH Lee 7 , WY Tak 8 , YJ Lee 9 , and K Yoo 1<br />

1 Ewha Womans University, Seoul, South Korea;<br />

2 Konkuk University Hospital, Seoul, South Korea; 3 Inje<br />

University Ilsan Paik Hospital, Gyeonggi-do, South Korea;<br />

4 Youngnam University Medical Center, Daegu, South<br />

Korea; 5 Kosin Medical School, Busan, South Korea; 6 Inje<br />

University Sanggye Paik Hospital, Seoul, South Korea;<br />

7 Sungkyunkwan University Samsung Medical Center,<br />

Seoul, South Korea; 8 Kyungpook National University<br />

Hospital, Daegu, South Korea; 9 Inje University Busan<br />

Paik Hospital, Busan, South Korea<br />

BACKGROUND: In the pivotal phase III clinical<br />

trials, clevudine 30 mg for 6 months showed<br />

potent antiviral activity and significant biochemical<br />

improvement along with a marked post-treatment<br />

antiviral effect. This analysis was performed to<br />

evaluate the early biochemical and virological<br />

response of clevudine in chronic hepatitis B patients<br />

with cirrhosis.<br />

METHODS: The data of 13 patients who were<br />

diagnosed chronic hepatitis B patients with cirrhosis<br />

were collected from 1 hospital. Preliminary results who<br />

have been treated for at least 1 month are presented<br />

here. HBV DNA was quantified by bDNA assay with a<br />

lower limit of detection of 141,500 copies/mL.<br />

RESULTS: Median HBV DNA levels before therapy<br />

was 7.3 log 10<br />

copies/mL and the median changes in<br />

HBV DNA levels from baseline was –1.5 log 10<br />

copies/<br />

mL after 1 month of therapy (n=13) and –2.7 log 10<br />

copies/mL after 3 month of therapy (n=4). Serum<br />

HBV DNA levels were below 141,500 copies/mL in<br />

46% (6/13) at 1 month and 75% (3/4) after 3 months<br />

of therapy.<br />

At baseline, overall median ALT was 51 IU/L and 3<br />

patients had normal ALT. Forty-six percent (46%,<br />

6/13) at month 1 and 100% (4/4) of patients at<br />

month 3 had normal ALT.<br />

CONCLUSION: Clevudine 30 mg once daily therapy<br />

demonstrated early viral suppression and significant<br />

biochemical improvement in patients with liver<br />

cirrhosis.<br />

Abstract 30<br />

Clevudine was Superior to<br />

Lamivudine in the Patients with<br />

HBeAg(+) Chronic Hepatitis B<br />

GK Lau 1 , N Leung 2 , CK Hui 1 , A Kwok 2 , A Wong 2 ,<br />

R Chan 2 , SG Hwang 3 , and HS Lee 4<br />

1 The University of Hong Kong, Hong Kong, PRC; 2 Alice<br />

Ho Miu Ling Nethersole Hospital, Hong Kong, PRC;<br />

3 Bukwang Pharmaceutical Co., Ltd., Seoul, Korea; 4 Seoul<br />

National University Hospital, Seoul, Korea<br />

BACKGROUND: Clevudine is a pyrimidine analogue<br />

with potent anti-HBV activity in vitro. In the<br />

pivotal phase III clinical trials, clevudine 30 mg for<br />

24 weeks showed profound viral suppression with<br />

normalization of serum ALT levels.<br />

METHODS: The aim of this study is to compare the<br />

efficacy and safety of clevudine versus lamivudine for<br />

48 weeks in chronic hepatitis B (CHB) patients in a<br />

randomized and blinded way. The study is ongoing<br />

at two sites in Hong Kong. Eligible patients were<br />

treatment-naïve HBeAg(+) CHB patients with HBV<br />

DNA levels ≥ 3 × 10 6 copies/mL and 1.0 ≤ALT


RESULTS: The clevudine group demonstrated greater<br />

viral suppression at Week 48 when compared with<br />

the Lamivudine group [median reduction (range)<br />

4.9 (3.3-6.2) vs. 3.2 (0.8-5.6) log 10<br />

copies/ml at Week<br />

48 respectively, p < 0.05]. Serum HBV DNA levels<br />

were below 300 copies/mL in 73% and 27% at week<br />

32 and in 82% and 36% at week 48 in the clevudine<br />

and lamivudine groups, respectively. Nine patients<br />

of clevudine group and 10 patients of lamivudine<br />

group had normal ALT at week 48 (p=0.82). In the<br />

lamivudine group, viral breakthrough occurred in 3<br />

patients during week 32-48, but no patient had viral<br />

breakthrough for 48 weeks in the clevudine group.<br />

Lamivudine-resistant mutations (rtL180M and<br />

rtM204V) were detected in all of the three patients<br />

with viral breakthrough in the lamivudine group.<br />

Clevudine was well tolerated with the incidence of<br />

adverse events comparable to lamivudine.<br />

CONCLUSIONS: A 48-week dosing with clevudine<br />

30mg showed superior viral suppression to<br />

lamivudine 100mg without the emergence of viral<br />

breakthrough in HBeAg(+) CHB patients, while 3<br />

in the 11 patients (27.3%) in the lamivudine group<br />

had viral breakthrough during 48 weeks which were<br />

associated with lamivudine-resistant mutations.<br />

Abstract 31<br />

Clevudine Monotherapy Showed<br />

Rapid Viral and Biochemical<br />

Response in Chronic Hepatitis B<br />

Patients with Cirrhosis<br />

CH Lee 1 , KW Chung 2 , JS Lee 3 , HJ Lee 4 , BH Han 5 ,<br />

WC Choi 6 , SW Paik 7 , KC Koh 7 , JH Lee 7 , WY Tak 8 ,<br />

YJ Lee 9 , and HY Lee 10<br />

1 Konkuk University Hospital, Seoul, South Korea;<br />

2 Ewha Womans University, Seoul, South Korea; 3 Inje<br />

University Ilsan Paik Hospital, Gyeonggi-do, South Korea;<br />

4 Youngnam University Medical Center, Daegu, South<br />

Korea; 5 Kosin Medical School, Busan, South Korea; 6 Inje<br />

University Sanggye Paik Hospital, Seoul, South Korea;<br />

7 Sungkyunkwan University Samsung Medical Center,<br />

Seoul, South Korea; 8 Kyungpook National University<br />

Hospital, Daegu, South Korea; 9 Inje University Busan<br />

Paik Hospital, Busan, South Korea; 10 Chungnam<br />

National University Hospital, Daejeon, South Korea<br />

BACKGROUND: In the pivotal phase III clinical trials,<br />

clevudine 30 mg for 6 months showed potent antiviral<br />

activity and significant biochemical improvement<br />

along with a marked post-treatment antiviral effect.<br />

This analysis was performed to evaluate the efficacy<br />

of clevudine in chronic hepatitis B patients with<br />

cirrhosis.<br />

METHODS: The data of 23 patients were collected<br />

from the hospitals. The patients were diagnosed<br />

chronic hepatitis B patients with hepatitis B-related<br />

cirrhosis. Among them, 5 patients were diagnosed with<br />

decompensated liver cirrhosis or HCC. Preliminary<br />

results from the 23 patients who have been treated<br />

for at least 12 weeks are presented here.<br />

RESULTS: Median HBV DNA levels before therapy<br />

was 6.03 log 10<br />

copies/mL (n=23). The median changes<br />

in HBV DNA levels from baseline was –3.43 log 10<br />

copies/mL (n=18) after 12-16 weeks of therapy and<br />

–3.50 log 10<br />

copies/mL (n=12) after 20-24 weeks of<br />

therapy. Serum HBV DNA levels were below 300<br />

copies/mL in 39 % (n=18) after 12-16 weeks of<br />

therapy and 75 % (n=12) after 20-24 weeks of therapy.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 31


From the 23 patients, 20 patients (87%) had HBV<br />

DNA


polymerase. The molecular mechanism of action of<br />

clevudine inhibition is not completely understood. In<br />

this study, we have examined the unique mechanism<br />

of action of clevudine.<br />

METHODS: An endogenous HBV polymerase assay<br />

was employed to investigate the mechanism of action<br />

of CLV-TP. The reaction was followed by measuring the<br />

incorporation of 32 P-labeled nucleoside triphosphates<br />

into HBV DNA using HBV core particles. In order<br />

to determine the mode of inhibition, reactions were<br />

followed in the presence of varying concentrations<br />

of CLV-TP and a natural nucleotide substrate. A<br />

control experiment was performed using a known<br />

competitive inhibitor, 3TC-TP.<br />

RESULTS: Since clevudine is a thymidine analog,<br />

inhibition of HBV DNA polymerase by CLV-TP<br />

was examined by varying both CLV-TP and TTP<br />

concentrations. The data were fit to both competitive<br />

and non-competitive equations. The plots clearly fit<br />

the non-competitive equation with the inhibition<br />

constant (K i<br />

) of 0.67 µM. The non-competitive<br />

inhibition of CLV-TP was confirmed by performing the<br />

same experiment in the presence fixed concentration<br />

of TTP and varying dCTP which should not compete<br />

with CLV-TP. A similar inhibition profile was observed<br />

with the K i<br />

of 1.15 µM. A control experiment<br />

was performed with 3TC-TP and as expected, it<br />

competitively inhibited HBV DNA polymerase with<br />

the K i<br />

of 0.0075 µM which was comparable to the<br />

published data.<br />

CONCLUSIONS: Using an endogenous HBV<br />

polymerase assay, the mode of inhibition for CLV-TP<br />

was determined to be non-competitive. Since several<br />

active site mutations are known to confer resistance<br />

to Clevudine in vitro, CLV-TP may be binding at or near<br />

the active site of the HBV polymerase without being<br />

utilized as a substrate. To the best of our knowledge<br />

this is the first example of a nucleoside analog<br />

triphosphate showing non-competitive inhibition.<br />

Abstract 34<br />

Mechanistic Characterization<br />

of Potent Small Molecule HCV<br />

Inhibitors that Target NS5A<br />

A Sandrasagra, AMI Lam, R Fathi, Z Yang, J Cao,<br />

Y Liu, G Li, Y Liao, Q Zhu, K Nawoschik, H-J Cho,<br />

R Wu, G Westby, and CR Wobbe<br />

XTL Biopharmaceuticals Inc, Valley Cottage, New York,<br />

USA<br />

Background: Target based screening has led to the<br />

discovery of a number of small molecule therapeutics<br />

that inhibit HCV by acting at enzymes critical for<br />

viral replication. Complimentary phenotypic replicon<br />

based screening approaches, which are un-biased<br />

toward a specific molecular target, can enable the<br />

identification of otherwise inaccessible and novel<br />

targets or pathways. We have employed replicon<br />

based screening to identify a novel series of small<br />

molecule HCV replication inhibitors that affect NS5A<br />

dependent functions.<br />

Methods and Results: We have generated potent<br />

HCV replicon inhibitors that have no toxicity against<br />

a range of cultured cell lines, and that induce a rapid<br />

multiple-log reduction of HCV RNA with no rebound<br />

upon inhibitor withdrawal. Our leads do not inhibit<br />

in vitro assays for HCV protease, helicase, polymerase<br />

and IRES. Consistent with these findings, replicons<br />

with resistance mutations to NS3 protease and NS5B<br />

polymerase inhibitors are susceptible to our leads.<br />

XTL leads were also found to be inactive vs. a number<br />

of cellular metabolic pathways and targets including<br />

nucleotide pools, caspases, a panel of 50 in vitro protein<br />

kinases assays, and a panel of 60 receptor binding<br />

assays. Combination studies with other known HCV<br />

replication inhibitors demonstrated that our leads<br />

are either additive or synergistic. Analysis of replicon<br />

mutations conferring resistance to our leads indicated<br />

that resistance could arise by the alteration of specific<br />

viral sequences outside of the NS3 protease or NS5B<br />

polymerase regions. Furthermore, a single mutation<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 33


(Y93H) within domain 1 of NS5A is sufficient to confer<br />

6 to 100 fold resistance vs. a panel of representative<br />

leads. Data from molecular genetic studies and in vitro<br />

binding assays suggest that XTL leads interact with<br />

NS5A. Affinity models constructed using the NS5A<br />

domain 1 crystal structure predict that our leads bind<br />

at or near the dimer interface and make contacts that<br />

are distinct from other compounds reported to target<br />

NS5A.<br />

Conclusion: Using a phenotypic replicon based<br />

screening approach in combination with Diversity<br />

Oriented Synthesis chemistries, we have generated<br />

potent HCV replication inhibitors with nanomolar<br />

EC 50<br />

s. Resistance selection, molecular genetic, in vitro<br />

binding, and molecular modeling studies suggest that<br />

our leads that act by interacting with NS5A.<br />

Abstract 35<br />

GSK949614; a Novel and Potent<br />

Thumb Site Inhibitor of the HCV<br />

NS5B Polymerase<br />

PA Thommes 1 , NR Parry 1 , EM Amphlett 1 , G Bravi 2 ,<br />

H Bright 1 , AG Cheasty 1 , MA Convery 2 , JA Corfield 1 ,<br />

R Fenwick 1 , DF Gray 1 , RM Grimes 1 , D Harrison 1 ,<br />

CD Hartley 4 , RL Jarvest 4 , KJ Medhurst 4 , ML Meeson 4 ,<br />

D Mesogiti 1 , F Mirzai 1 , JE Mordaunt 4 , NA Roughley 3 ,<br />

P Shah 4 , MJ Slater 4 , JH Thorpe 2 , CS Wilkinson 1 ,<br />

and E Williams 1<br />

1 Infectious Diseases CEDD, Stevenage, 2 Computational<br />

& Structural Chemistry, GSK Medicines Research<br />

Centre, Gunnels Wood Road, Stevenage SG1 2NY, United<br />

Kingdom<br />

Background: The HCV NS5B gene encodes the<br />

viral RNA-dependent RNA polymerase which is<br />

essential for HCV replication and an attractive<br />

target for antiviral therapy. In common with other<br />

polymerases, the HCV polymerase has a structure<br />

similar to a right hand, with distinguishable ‘finger’,<br />

‘palm’ and ‘thumb’ domains. Several classes of<br />

inhibitors directed against HCV NS5B have been<br />

described, which inhibit both enzymatic activity and<br />

replication. These compounds bind to distinctive<br />

sites on the polymerase molecule and thus have been<br />

termed ‘palm’, ‘thumb’, ‘finger-loop’ binders, etc.<br />

We have previously reported the identification of<br />

two distinct series with different binding modes in<br />

the palm site of the polymerase; thiadiazines and<br />

acylpyrrolidines. Here we describe a pyrazole based<br />

inhibitor series which binds at the thumb site of the<br />

enzyme.<br />

Methods: The development of the pyrazole series<br />

was driven by structure-based design with parallel<br />

use of NS5B enzyme and HCV genotype 1a and 1b<br />

replicon assays and by monitoring PK parameters of<br />

key compounds in the rat. Resistant mutants were<br />

selected by growing replicon cells in the presence<br />

of compound and individual mutations assessed in<br />

enzyme and transient replicon assays. Selectivity<br />

was measured against the polymerases of other<br />

members of the Flaviviridae and human cellular DNA<br />

polymerases. Cytotoxicity was determined in Vero<br />

cells.<br />

Results: Using structure activity relationship driven<br />

by potency in in vitro assays the pyrazole series was<br />

optimised to provide GSK949614 as a lead molecule.<br />

This compound was shown to be a selective inhibitor<br />

of the HCV polymerase with sub-micromolar potency<br />

against HCV genotypes 1a and 1b in both enzyme<br />

and replicon assays. Its activity is synergistic with<br />

that of interferon alpha 2a in replicon assays.<br />

GSK949614 has little cytotoxicity and no significant<br />

activity against either human DNA polymerases or<br />

the polymerases of related viruses. Replicon cells<br />

grown in the presence of GSK949614 gave rise to<br />

mutations at amino acids 423 and 419, located in the<br />

thumb region of the enzyme. Each of these mutations<br />

was shown to be critical for the sensitivity of the<br />

enzyme to the inhibitor. The compound retains full<br />

activity against a panel of enzymes resistant to drugs<br />

binding to other sites on the polymerase, including<br />

the palm site binders. GSK949614 has bioavailability<br />

in both rat and dog that will allow extended safety<br />

assessments.<br />

34 Global Antiviral Journal Volume 3, Supplement 2


Conclusion: We have developed the pyrazole series<br />

binding in the thumb region of the NS5B molecule<br />

to yield GSK949614. Its in vitro properties, together<br />

with its PK profile, make it suitable for further study<br />

as a potential drug candidate.<br />

Abstract 36<br />

Pyrrolo[1,2-b]pyridazin-2-ones<br />

Show Promising In Vitro Antiviral<br />

Activity Against HCV NS5B<br />

Polymerase<br />

F Ruebsam 1 , SE Webber 1 , MT Tran 1 , CV Tran 1 ,<br />

DE Murphy 1 , J Zhao 2 , PS Dragovich 1 , SH Kim 3 ,<br />

L Li 4 , Y Zhou 1 , Q Zhao 1 , CR Kissinger 1 , RE Showalter 1 ,<br />

M Lardy 1 , A Shah 5 , M Tsan 6 , R Patel 1 , L LeBrun 1 ,<br />

R Kamran 7 , MV Sergeeva 1 , and L Kirkovsky 1<br />

1 Anadys Pharmaceuticals, Inc., 3115 Merryfield Row,<br />

San Diego, CA 92121, USA; 2 Celgene Corporation, 4550<br />

Towne Centre Court, San Diego, CA 92121, USA; 3 Arena<br />

Pharmaceuticals, Inc., 6166 Nancy Ridge Drive, San<br />

Diego, CA 92121, USA; 4 Intellikine, Inc., 10931 North<br />

Torrey Pines Road, Suite 103, La Jolla, CA 92037, USA;<br />

5 SGX Pharmaceuticals, Inc., 10505 Roselle Street, San<br />

Diego, CA 92121, USA; 6 Illumina, Inc., 9885 Towne<br />

Centre Drive, San Diego, CA 92121, USA; 7 Takeda San<br />

Diego, 10410 Science Center Drive, San Diego, CA 92121,<br />

USA<br />

Background: Hepatitis C virus (HCV) is a major<br />

cause of acute hepatitis and chronic liver disease,<br />

including cirrhosis and liver cancer. Low response<br />

rates to the current standard of care, in particular<br />

for patients infected with genotype 1 HCV, along<br />

with significant side-effects of current HCV therapy<br />

result in a continuing medical need for improved<br />

treatments.<br />

Methods: As part of our efforts to identify novel<br />

small molecule, non-nucleoside inhibitors of the HCV<br />

NS5B protein using a structure based design approach,<br />

we investigated a series of pyrrolo[1,2-b]pyridazin-2-<br />

ones (Figure 1). We systematically explored variation<br />

of the substituents at the 1-, 6- and 7’-positions of<br />

the pyrrolo[1,2-b]pyridazin-2-ones. We also assessed<br />

the biological properties of the resulting molecules<br />

including inhibition of the 1b NS5B enzyme, activity<br />

in the HCV genotype 1b subgenomic replicon in tissue<br />

culture, cytotoxicity, and stability toward human liver<br />

microsomes (HLM).<br />

Results: Described here are the structure-activity<br />

relationships observed by varying the 1-, 6- and 7’-<br />

substituents of the pyrrolo[1,2-b]pyridazin-2-one<br />

NS5B inhibitors under study. We observed that while<br />

certain alkyl groups in R 2 , such as isoamyl or tertbutyl<br />

ethyl fragments, led to compounds with low<br />

nanomolar potencies in biochemical and cell-based<br />

assays, the introduction of a 4-fluorobenzyl moiety<br />

provided the best overall balance between potency<br />

and metabolic stability. We also noted that the R 3<br />

position was very sensitive to structural changes and<br />

that a sulfonamide moiety was critical for activity.<br />

The combination of optimal substituents at positions<br />

1, 6 and 7’ resulted in a compound that demonstrated<br />

potent inhibition against the 1b NS5B enzyme (IC 50<br />


Abstract 37<br />

4-(1,1-Dioxo-1,4-dihydro-1λ 6 -<br />

benzo[1,4]thiazin-3-yl)-5-<br />

hydroxy-2H-pyridazin-3-ones are<br />

a Novel Series of Molecules which<br />

Inhibit the HCV NS5B Polymerase<br />

CV Tran 1 , DA Ellis 2 , J Blazel 3 , PS Dragovich 1 , Z Sun 4 ,<br />

F Ruebsam 1 , HM McGuire 5 , AX Xiang 1 , J Zhao 6 ,<br />

L Li 7 , Y Zhou 1 , SE Webber 1 , Q Han 1 , CR Kissinger 1 ,<br />

RE Showalter 1 , M Lardy 1 , A Shah 2 , M Tsan 8 , R Patel 1 ,<br />

L LeBrun 1 , R Kamran 9 , MV Sergeeva 1 , and L Kirkovsky 1<br />

1 Anadys Pharmaceuticals, Inc., 3115 Merryfield Row,<br />

San Diego, CA 92121, USA; 2 SGX Pharmaceuticals, Inc.,<br />

10505 Roselle Street, San Diego, CA 92121, USA;<br />

3 Ardea Biosciences, Inc., 3300 Hyland Avenue, Costa<br />

Mesa, CA 92626, USA; 4 Merck & Co., Inc., 126 E.<br />

Lincoln Avenue, Rahway, NJ 07065, USA; 5 AstraZeneca<br />

Pharmaceuticals LP, 35 Gatehouse Drive, Waltham, MA<br />

02451, USA; 6 Celgene Corporation, 4550 Towne Centre<br />

Court, San Diego, CA 92121, USA; 7 Intellikine, Inc.,<br />

10931 North Torrey Pines Road, Suite 103, La Jolla, CA<br />

92037, USA; 8 Illumina, Inc., 9885 Towne Centre Drive,<br />

San Diego, CA 92121, USA; 9 Takeda San Diego, 10410<br />

Science Center Drive, San Diego, CA 92121, USA<br />

Background: Hepatitis C virus (HCV) is a major<br />

cause of acute hepatitis and chronic liver disease,<br />

including cirrhosis and liver cancer. Globally, an<br />

estimated 170 million individuals, 3% of the world’s<br />

population, are chronically infected with HCV and<br />

3 to 4 million people are newly infected each year.<br />

Currently, there is no vaccine available to prevent<br />

hepatitis C, nor a HCV-specific antiviral agent<br />

approved for treatment of chronic hepatitis C. The<br />

current standard of care is a combination of pegylated<br />

interferon (IFN) with ribavirin. However, response<br />

rates in patients infected with genotype 1 HCV,<br />

along with adverse side-effects, result in a continuing<br />

medical need for improved therapy.<br />

determined using the NS5B 1b enzyme. EC 50<br />

and<br />

cytotoxicity (CC 50<br />

) values were generated using a Huh-7<br />

HCV replicon cell line. Metabolic stability studies<br />

were conducted using human liver microsomes.<br />

Results: The newly discovered class of molecules,<br />

4-(1,1-dioxo-1,4-dihydro-1λ 6 -benzo[1,4]thiazin-3-<br />

yl)-5-hydroxy-2H-pyridazin-3-ones, showed excellent<br />

in vitro characteristics. The majority of compounds<br />

in this series were generally not cytotoxic (CC 50<br />

),<br />

had moderate to high stability towards human liver<br />

microsomes (HLM) and were potent inhibitors of<br />

both the NS5B polymerase enzyme (IC 50<br />

< 18µM)<br />

and the HCV replicon (EC 50<br />

< 1µM). One optimized<br />

compound exhibited an IC 50<br />

< 10 nM against the 1b<br />

NS5B enzyme and an EC 50<br />

of 1.1 nM when tested<br />

against the 1b HCV replicon in cell culture. This<br />

compound was also moderately stable in the presence<br />

of HLM with a half-life of 32 minutes.<br />

Conclusion: The novel series of 4-(1,1-dioxo-1,4-<br />

dihydro-1λ 6 -benzo[1,4]thiazin-3-yl)-5-hydroxy-2Hpyridazin-3-ones<br />

(shown in Figure 1) displayed an<br />

excellent in vitro profile against HCV NS5B polymerase.<br />

The inhibitors had low nanomolar activity against<br />

the NS5B enzyme as well as HCV replicon, and<br />

were reasonably stable in the presence of HLM. The<br />

encouraging in vitro data support further preclinical<br />

evaluation of this series in order to confirm its utility<br />

for the treatment of chronic HCV infection.<br />

O O<br />

S<br />

OH<br />

R 1<br />

N<br />

H<br />

N N O<br />

R 2<br />

Figure 1<br />

R 3<br />

Methods: Using a structure based design approach,<br />

we identified and optimized a novel series of<br />

compounds that inhibit HCV NS5B polymerase<br />

with excellent in vitro properties. IC 50<br />

values were<br />

36 Global Antiviral Journal Volume 3, Supplement 2


Abstract 38<br />

Interference of Hsp90 Activity<br />

by Hsp90 Inhibitor Suppresses<br />

Hepatitis C Virus (HCV)<br />

Replication<br />

S Ujino 1 , S Yamaguchi 1 , K Shimotohono 3 , and<br />

H Takaku 1,2<br />

1 Department of Life and Environmental Sciences; 2 High<br />

Technology Research Center; 3 Research Institute, Chiba<br />

Institute of Technology, Chiba, Japan<br />

its inhibitory effects were caused by NS3 degradation.<br />

Moreover, as a result of immunoprecipitation,<br />

interaction between Hsp90 and NS3 was detected.<br />

CONCLUSIONS: Here, we present a new and potent<br />

strategy for therapy of hepatitis C virus (HCV)<br />

infection with Hsp90 inhibitor. We found that Hsp90<br />

inhibitor has an anti HCV activity. Furthermore,<br />

Hsp90 inhibitor was elucidated as non-escape drug by<br />

long-term treatment. HCV NS3 was found to mediate<br />

HCV replication after interaction that it is interacted<br />

with Hsp90. These results illustrate implications<br />

for the HCV replication cycle and novel antiviral<br />

strategies.<br />

BACKGROUND: The Hsps induced by cells stress are<br />

expressed at high levels in a wide range of tumors<br />

and are closely associated with a poor prognosis and<br />

resistant therapy. The principal folding protein belong<br />

to the Hsp70 and Hsp90, which bind to unfolded<br />

sequences in polypeptide substrates and shown for<br />

hydrophobic regions. As numerous oncoproteins<br />

have been shown to be Hsp90 client proteins, Hsp90<br />

inhibitors have become a arrest the growth of tumor<br />

cells. Previously we demonstrated the inhibition of<br />

HCV RNA replication in the HCV replicon with Hsp90<br />

inhibitor.<br />

METHODS: We used two types of HCV replicon cells.<br />

One is HCV full genome replicon(NNC) and the other<br />

is HCV subgenomic reilicon. HCV inhibiting effect was<br />

measured by Real-Time PCR. Western blot analysis of<br />

core and NS protein expression in NNC cells treated<br />

with 17-AAG. Lysates were immunoblotted with core<br />

and NS specific antibodies. Immunoprecipitation<br />

of Hsp90 and NS3 in cultured cells. NS3 was<br />

immunoprecipitated from cell lysates with anti-FLAG<br />

or anti-Hsp90 antibodies and immunoblotted by<br />

using respective antibodies as indicated.<br />

RESULTS: Replication of HCV was inhibited when<br />

treated with Hsp90-inhibitor(0.25~250nM). Hsp90<br />

inhibitor showed neither cytotoxicity nor virus escape<br />

drug in long-term culture assay. We also found that<br />

Hsp90 inhibited the formation of the NS3–Hsp90<br />

chaperone complex during HCV replication, and that<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 37


Hepatitis B Foundation Symposium:<br />

Early Detection and<br />

Management of Cirrhosis and<br />

Hepatocellular Carcinoma<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 39


Abstract 39<br />

Hepatocellular Carcinoma: Why<br />

Early Diagnosis is Needed<br />

AM Di Bisceglie<br />

Saint Louis University Liver Center, Saint Louis, MO, USA<br />

Hepatocellular carcinoma (HCC) is one of the frequent<br />

solid malignancies world-wide and is increasing in<br />

incidence in the United States and developed western<br />

world. The occurrence of HCC is very closely linked<br />

to the presence of underlying chronic liver disease,<br />

including chronic viral hepatitis and cirrhosis. In the<br />

United States, the frequency of various underlying<br />

diseases varies regionally but overall, chronic viral<br />

hepatitis accounts for 50 to 60% of all cases, while<br />

cirrhosis due to alcohol and other conditions accounts<br />

for another substantial proportion.<br />

Although it has been shown that early diagnosis of<br />

HCC is possible, most patients seem to present when<br />

this tumor is at an advanced stage. Typical symptoms<br />

that lead to the diagnosis include abdominal pain,<br />

weight loss and further hepatic decompensation in<br />

someone known to have cirrhosis. Unfortunately,<br />

when such symptoms are present, the tumor is often<br />

very large and may even be metastatic, either within<br />

or beyond the liver. At this stage, the tumor is rarely,<br />

if ever, respectable and there are limited potentially<br />

curative therapeutic options available for the patient.<br />

Liver transplantation is associated with optimal<br />

results when the tumor is within the so-called “Milan<br />

criteria” – that is, a single tumor 3cm in diameter. Local ablative treatments such<br />

as ethanol injection or radio-frequency ablation are<br />

similarly most effective with tumor diameters


Serological tests have the ability to correctly define<br />

fibrosis stage as either mild (F0/F1) or significant<br />

(F2-4) in up to 80% of patients with some indices<br />

better at predicting advanced disease and others at<br />

excluding advanced disease.<br />

Serological tests are accurate enough to abrogate the<br />

need for liver biopsy in 40% of patients.<br />

References:<br />

Bedossa P, et al, Hepatology 2003;38:1449-57.<br />

Cales P, et al, Hepatology 2005;42:1373-81.<br />

Imbert-Bismut F, et all, Lancet 2001;357:1069-75.<br />

Adams LA, et al, Clin Chem 2005;51:1867-73.<br />

Patel K, et al, J Hepatol 2004;41:935-42.<br />

Ziol M, et al, Hepatology 2005;41:48-54.<br />

Castera L, et al, Gastroenterology 2005;128:343-50.<br />

Tests of liver stiffness with elastography are able<br />

to predict cirrhosis in 90% of patients and can be<br />

complimentary to blood tests.<br />

Algorithms of combination of biopsy, serological<br />

tests and elastography may be the optimal way to<br />

stage and monitor fibrosis.<br />

Table: Serological Tests for Liver Fibrosis<br />

Patients<br />

Name (Serum Markers)<br />

AUROC<br />

(95% CI)<br />

Sens Spec PPV NPV<br />

Wai et al (2003) 192<br />

APRI<br />

(AST, platelets)<br />

0.88<br />

(.80-.96)<br />

41% 95% 88% 64%<br />

Rosenberg et al (2004) 1021<br />

ELF<br />

(Propeptide III collagen, TIMP 1,<br />

HA)<br />

0.80<br />

(.76-.85)<br />

90.5% 41% 99% 92%<br />

Ziol et al<br />

(2005)<br />

327<br />

Fibroscan<br />

(hepatic elastography)<br />

0.79<br />

(.73-.84)<br />

56 % 91% 88% 56%<br />

Imbert-Bismut et al<br />

(2001)<br />

339<br />

Fibrotest<br />

(α 2<br />

macroglobulin, α 2<br />

globulin, γ<br />

globulin, apolipoprotein A 1<br />

, γGT<br />

and total bilirubin)<br />

0.87<br />

(SD<br />

0.34)<br />

87 % 59% 63% 85%<br />

Castera et al (2005) 183<br />

Combined Fibroscan and<br />

Fibrotest<br />

0.88<br />

(.82-.92)<br />

NA NA NA NA<br />

Patel et al<br />

(2004)<br />

402<br />

Fibrospect<br />

hyaluronic acid, tissue inhibitor of<br />

metalloproteinase 1 (TIMP-1) and<br />

α 2<br />

macroglobulin<br />

0.831 77% 73% 74% 76%<br />

Adams et al. (2005) 221<br />

Hepascore<br />

Bilirubin, γGT, hyaluronic acid, α 2<br />

macroglobulin, age and sex<br />

0.82<br />

0.90<br />

0.89<br />

63%<br />

88%<br />

71%<br />

89%<br />

74%<br />

89%<br />

88%<br />

88%<br />

95%<br />

98%<br />

98%<br />

42 Global Antiviral Journal Volume 3, Supplement 2


Abstract 41<br />

Screening Fibrosis: “La Révolution<br />

Française”<br />

T Poynard<br />

Groupe Hospitalier Pitié-Salpêtrière, France<br />

Non-invasive tests, biomarkers and liver stiffness<br />

measurements (LSM) have advantages over other<br />

proposed strategies for liver injury assessment. Several<br />

patented biomarkers are already on the market, the<br />

most validated being FibroTest (FT). FT has similar<br />

diagnostic value in the most frequent chronic liver<br />

diseases, and at least similar false positive or false<br />

negative rates than routine biopsy (Poynard, BMC<br />

Gastroenterology 2007).<br />

Practices are evolving rapidly and in France 81% of<br />

hepatologists used FT and 32% used LSM (Castera, J<br />

Hepatol 2007).<br />

French health authorities concluded that FT was<br />

a valid alternative to biopsy in chronic hepatitis C<br />

and the reimbursement is pending (http://www.hassante.<br />

fr/portail/display.jsp?id=c_476486).<br />

We conducted three fibrosis screening studies. The<br />

first study (FibroGras) analyzed retrospectively<br />

the frozen sera of a consecutive cohort of 1,909<br />

hyperlipidaemic patients prospectively in followed<br />

a lipid centre. A total of 925 blood donors have<br />

been taken as controls (Ctl). Advanced fibrosis was<br />

presumed by FT in 2.8% hyperlipidaemic patients<br />

vs. 0% in Ctl (P < 0.0001); advanced steatosis was<br />

presumed by SteatoTest in 30.1% vs. 4.9% Ctl (P <<br />

0.0001) and NASH was presumed by NashTest in<br />

7% vs. 0%, respectively (P < 0.0001). There was a<br />

highly significant and linear association between<br />

the number of metabolic syndrome factors and liver<br />

disease prevalence presumed with biomarkers – the<br />

highest being for type 2 diabetics: advanced steatosis<br />

66%, NASH 24% and advanced fibrosis 6% (Ratziu,<br />

Aliment Pharmacol Ther 2007).<br />

The second study (FibroSucre) analyzed prospectively<br />

1,131 consecutive patients, without a history of<br />

liver disease, followed in a diabetes center. The<br />

biomarker predicted advanced fibrosis in 5.6%. A<br />

total of 45 patients with presumed advanced fibrosis<br />

were re-investigated using LSM and standard tests,<br />

and advanced fibrosis was confirmed in 32 patients<br />

(2.8%), with 5 cases of cirrhosis including 4 cases with<br />

of hepatocellular carcinoma. Most of liver diseases<br />

previously unknown were NASH but interestingly in<br />

2 patients a chronic hepatitis C has been discovered.<br />

In the population with type 2 diabetes, 45 years or<br />

older, the prevalence of confirmed advanced fibrosis<br />

was 4.3% and hepatocellular carcinoma was 5.7/1,000<br />

(Jacqueminet, J Hepatol 2006;44:A260).<br />

The third study (FibroCPAM) is on going in a general<br />

population. Consecutive apparently healthy subjects<br />

>40 years had free health check-up offered by social<br />

security centres. Subjects with presumed advanced<br />

fibrosis at FT, were re-investigated by a hepatologist<br />

using LSM, and if necessary, ultrasonography,<br />

endoscopy or liver biopsy. A scheduled intermediate<br />

analysis was performed in the first 5,013 subjects.<br />

FT predicted advanced fibrosis in 3%, including 0.4%<br />

cirrhosis. At the time of abstract submission 40%<br />

of subjects with presumed advanced fibrosis have<br />

been fully reinvestigated. Fibrosis was confirmed in<br />

50%, mostly with LSM >8.8kPa including 8 cases of<br />

cirrhosis previously unknown. As expected in France,<br />

most of liver diseases discovered were NASH and<br />

alcoholic liver diseases, but 5 previously unknown<br />

hepatitis C and one hepatitis B were also identified.<br />

Finally in a world without perfect Gold Standard,<br />

we developed in 2,004 patients a new non-invasive<br />

methodology for improving accuracy of fibrosis<br />

markers using the strength of concordance between<br />

FT and LSM (Poynard, Hepatology 2007;46:1071).<br />

According to the respective risk of false negative/<br />

positive, the present results strongly suggest for<br />

fibrosis screening, to use FT at first line and LSM at<br />

second line. Patients not concordant at second line<br />

must be re-investigated by repeated biomarkers or<br />

liver biopsy.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 43


In conclusion, waiting for an ideal non-invasive<br />

biomarker is a dream, and it is time to screen for<br />

advanced fibrosis.<br />

Abstract 42<br />

Treatment of HCC Over the Past<br />

Decade: The Experience of Over<br />

Four Thousand Patients in Japan<br />

M Omata<br />

Department of Gastroenterology, University of Tokyo<br />

Etiology: Approximately 34,000 patients died<br />

of hepatocellular carcinoma last year in Japan. Of<br />

these, 11% and 83% of the patients with the cancer<br />

were positive for HBV and for HCV, respectively. This<br />

indicates that 94% of our patients with hepatocellular<br />

carcinoma are currently infected with either of two<br />

hepatitis viruses. In contrast, only 3% (1.5% for HBV<br />

and 1.5% for HCV) of general population are infected<br />

with the viruses.<br />

Natural Course: Our follow-up study indicates<br />

that there is difference between B-viral and C-viral<br />

disease to develop into hepatocellular carcinoma, e.g.,<br />

C-viral hepatocellular carcinoma often develop with<br />

the background of advanced fibrosis and/or cirrhosis,<br />

whereas B-viral HCC sometimes without. Therefore,<br />

surgical resection or complete ablation of nodules not<br />

necessary leads to complete cure of hepatocellular<br />

carcinoma, especially in C-viral HCCs.<br />

Our Treatment Strategy: In fact, our<br />

experience of more than 5000 patients treated by PEIT<br />

(Percutaneous Ethanol Injection Therapy), PMCT<br />

(Percutaneous Microwave Coagulation Therapy) and<br />

recent RFA (Radio Frequency Ablation), indicates<br />

frequent recurrence. It is clear that you need the<br />

treatment both for backgrounds and tumor nodules.<br />

Otherwise, the recurrence of cancer from background<br />

(cirrhotic nodules) could reach 80% within 5 years.<br />

Strategy of ours is to treat cancer nodules by<br />

PEIT, PMCT and RFA. Approximately 85% of our<br />

patients who are admitted to our Department of<br />

Gastroenterology, University of Tokyo, were treated<br />

with one of the above percutaneous methods.<br />

Recently, we have completed a prospective<br />

controlled study of PEIT and RFA (Gastroenterology<br />

2005;129:122-130). In that study, the RFA treated<br />

patients’ survival were significantly better than<br />

those of PEIT. Thus, majority of the patients are now<br />

treated by RFA , resulting in 3000 cases so far and<br />

only exceptionally cases are treated by PEIT.<br />

Recurrence: However, there are several problems.<br />

The biggest is the recurrence from cirrhotic background.<br />

Treatment for the backgrounds is basically to<br />

cure cirrhosis or advanced fibrosis. We have indicated<br />

that eradication of HCV by interferon eventually<br />

induces the resolution of fibrosis (Ann Intern Med<br />

2000;132:517-524) . In fact, this reduction of the<br />

fibrosis due to the interferon treatment were related<br />

to decrease of incidence of hepatocellular carcinoma<br />

(Ann Intern Med 1999;131:174-181). We initiated a<br />

prospective controlled study for the patients who had<br />

hepatocellular carcinoma, treated by percutaneous<br />

injection therapy and interferon. The result indicates<br />

that 21 patients treated by ablation and interferon<br />

which induced good response, 5-year survival were<br />

83%, compatible with liver transplantation (Ann<br />

Intern Med 2003;138:299-306).<br />

Advanced HCC: We still have many patients<br />

suffering from advanced hepatocellular carcinoma,<br />

especially with portal vein tumor invasion (PVI)<br />

who usually live only for 6 months. The combination<br />

of 5-FU and Interferon was given to more than 300<br />

patients with 17% of CR (Complete Response) (Cancer<br />

2006;106:1990-1997). Furthermore, the significance<br />

and the limitation of molecular targeting drugs on<br />

advanced cases could be shown in our patients.<br />

44 Global Antiviral Journal Volume 3, Supplement 2


Abstract 43<br />

Use of Pre-emptive Nucleoside<br />

Analogue Therapy for Hepatitis B<br />

Reactivation after Chemotherapy<br />

GKK Lau<br />

Queen Mary Hospital, The University of Hong Kong,<br />

Hong Kong SAR, China<br />

In areasc where hepatitis B infection is endemic,<br />

hepatitis due to hepatitis B virus reactivation after<br />

chemotherapy or immunosuppressive therapy,<br />

is a serious cause of liver-related morbidity<br />

and mortality. With the characterization of the<br />

underlying pathogenesis, much progress in the<br />

management of this important clinical problem has<br />

been made in the past 2 decades. By year 2007, it is<br />

mandatory to screen for hepatitis B surface antigen<br />

status before initiating intensive chemotherapy<br />

or immunosuppressive therapy. All those who are<br />

hepatitis B surface antigen positive should be started<br />

pre-emptive nucleos(t)ide analogues. However, there<br />

remains important issues, such as the type and length<br />

of nucleos(t)ide analogues therapy. As not all hepatitis<br />

B surface antigen positive patients will suffer from<br />

HBV reactivation, it will be useful to identify risk<br />

factors related to HBV reactivation so that patients<br />

will not be treated unnecessary with nucleos(t)ide<br />

analogues. In addition, there is an increase awareness<br />

of reactivation of occult hepatitis B virus, especially<br />

in hepatitis B virus endemic area, such as Asiapacific<br />

region. Careful epidemiological study will be<br />

needed to clarify the impact of occult hepatitis B<br />

infection in patients treated with chemotherapy or<br />

immunosuppressive therapy.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 45


Immunology & Pathogenesis<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 47


Abstract 44<br />

Cellular Immune Responses<br />

Against Hepatitis C in Acute and<br />

Chronic Infection<br />

MJ Koziel<br />

Beth Israel Deaconess Medical Center, Harvard Medical<br />

School, Boston MA, USA<br />

Hepatitis C virus (HCV) induces persistent infection<br />

and causes chronic liver diseases in most infected<br />

patients; however, the mechanisms whereby HCV<br />

evades an effective immune response and the role<br />

of immune responses in liver damage once chronic<br />

infection is established are unknown. Vigorous<br />

HCV-specific CD4+ and CD8+ T cell (CTL) responses<br />

against HCV multiple epitopes are necessary for<br />

spontaneous viral clearance during the acute phase,<br />

but the virus appears to have multiple strategies to<br />

evade these defenses. These include interference with<br />

the endogenous interferon response, attenuation and<br />

alteration of cellular function, and mutation to avoid<br />

immune recognition. Based on prospective treatment<br />

studies, it appears that the likelihood of establishing<br />

chronic infection is set as early as 12 weeks into<br />

infection. T cell responses may be associated with the<br />

likelihood of sustained virologic response in interferon<br />

and ribaviirn based regimens, but there is a relative<br />

paucity of data whether they are required for SVR.<br />

Despite an abundance of studies determining the<br />

correlates of protective immunity, there are relatively<br />

few studies on the role of immune responses during<br />

the chronic phase of infection. HCV-specific immune<br />

responses measured in the peripheral blood are weak<br />

and often barely detectable, whereas they can be<br />

found in liver tissue. CD4+ T cell responses appear<br />

to protect against liver injury and may be important<br />

to clearance during interferon and ribavirin based<br />

therapy. The classic understanding of CD8+ cells is<br />

that they are primarily involved in tissue injury, but<br />

there may be subpopulations of T cells that protect<br />

against liver inflammation.<br />

Abstract 45<br />

Broadly Neutralizing Antibodies<br />

to HCV: Definition of Epitopes and<br />

Antiviral Activity In Vitro and<br />

In Vivo<br />

M Law 1 , T Maruyama 1 , J Lewis 2 , E Giang 1 , AW Tarr 3 ,<br />

Z Stamataki 4 , P Gastaminza 1 , FV Chisari 1 , IM Jones 5 ,<br />

RI Fox 6 , JK Ball 3 , JA McKeating 4 , NM Kneteman 2 , and<br />

DR Burton 1<br />

1 The Scripps Research Institute, California, USA;<br />

2 University of Alberta, Alberta, Canada; 3 The University<br />

of Nottingham, Nottingham, UK; 4 University of<br />

Birmingham, Birmingham, UK; 5 University of Reading,<br />

Reading, UK; 6 Scripps Memorial Hospital, California,<br />

USA<br />

BACKGROUND: A major problem facing HCV vaccine<br />

design and attempts to prevent HCV transmission by<br />

passive antibody therapy, e.g. in the setting of liver<br />

transplantation, is the extreme variability of the virus.<br />

Indeed, several antibody preparations, including<br />

hepatitis C immune globulin (HCIG) prepared from<br />

human plasma pools, have been shown unable to<br />

protect against challenge with HCV quasispecies<br />

in animal models, or to prevent recurrence of HCV<br />

following transplantation. Key questions are whether<br />

neutralizing antibodies of sufficient breadth can be<br />

generated to protect against quasispecies challenge<br />

and whether the corresponding epitopes can be<br />

defined to facilitate vaccine design.<br />

METHODS: To investigate these questions, a<br />

phage-display antibody (Fab fragment) library was<br />

constructed using bone marrow RNA from a donor<br />

with chronic HCV infection and panned against<br />

HCV E1E2 envelope glycoprotein antigens to isolate<br />

specific human monoclonal antibodies (MAbs).<br />

Isolated Fabs with unique binding properties were<br />

converted into full length IgG1 molecules and their<br />

activity against a panel of diverse HCV isolates was<br />

examined to identify broadly neutralizing MAbs. The<br />

conserved neutralizing epitopes were mapped by<br />

competition with well-defined MAbs and by alanine<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 49


mutagenesis scanning. The correlation between in<br />

vitro neutralization and in vivo protection of the<br />

broadly neutralizing MAbs was investigated in passive<br />

antibody transfer experiments using a human liverchimeric<br />

mouse model.<br />

RESULTS: A panel of 36 distinct human Fabs specific<br />

for the HCV E2 envelope protein was isolated to define<br />

three antigenic regions (ARs) in E2. Seven distinct<br />

Fabs were converted to full length IgG1s to facilitate<br />

the characterization of the ARs; a series of in vitro<br />

assays showed that AR1 and AR3 partially overlap<br />

with the putative CD81 binding site but only AR3 is<br />

a target for multiple cross-neutralizing antibodies.<br />

Alanine mutagenesis scanning of regions Q412-S424,<br />

R483-P491 and G523-F550 of E2 identified shared<br />

and distinct residues used by CD81 and the MAbs,<br />

providing an explanation for such overlaps. Two<br />

AR3-antibodies (AR3A and AR3B), when given at a<br />

high dose (200 mg/kg animal), protected the human<br />

liver-chimeric mice against intravenous challenge<br />

with a heterologous HCV-infected human serum<br />

(2 x 10 5 IU, genotype 1a). While all 4 control mice were<br />

infected; AR3A antibody delayed virus replication and<br />

protected 2 of 5 mice while AR3B antibody protected<br />

3 of 4 mice.<br />

CONCLUSIONS: From a large panel of phage-display<br />

recombinant antibodies to HCV, we identified human<br />

MAbs that recognize a conserved antigenic region<br />

on the virus envelope glycoprotein E2, neutralize<br />

genetically diverse HCV isolates and protect against<br />

heterologous HCV quasispecies challenge in vivo in<br />

a human liver-chimeric mouse model. The results<br />

provide evidence that broadly neutralizing human<br />

antibodies to HCV protect against viral infection and<br />

suggest that a prophylactic vaccine against HCV may<br />

be achievable.<br />

Abstract 46<br />

Innate Immune Studies in<br />

Hepatitis B: Novel Studies in<br />

Pathogenesis and Treatment<br />

K Visvanathan<br />

Monash Institute of Medical Research, Australia<br />

Background/Aims: Toll-like receptors (TLR’s)<br />

are critical receptors that promote innate immune<br />

responses to pathogen-associated molecular<br />

patterns. Activation of TLR’s leads to production of<br />

cytokines such as TNF and IL-6. We have previously<br />

demonstrated that patients with HBeAg+ve CHB have<br />

suppressed TLR2 expression on peripheral monocytes<br />

and hepatocytes associated with decreased cytokine<br />

expression.<br />

Methods and Patients: Three different cohorts<br />

of patients were used for these studies. For the LMV<br />

studies, PBMCs was measured from 21 patients with<br />

untreated HBeAg-positive and HBeAg-ve CHB and 10<br />

uninfected control patients. Individual patient blood<br />

was stimulated with specific TLR ligands and the<br />

resultant supernatant was assayed for TNF and IL-6.<br />

In addition serum HBeAg was measured by enzyme<br />

immunoassay. For the IFN studies 15 HBeAg+ve<br />

patients all treated with IFN were used. Similar<br />

experiments to those for the LMV study were done<br />

for this study.<br />

Using a third cohort and using novel flow cytometric<br />

techniques we have further investigated the ability of<br />

hepatocytes and Kupffer cells from HBV patients to<br />

respond to culture with specific TLR stimuli. Primary<br />

human liver cells were cultured either unstimulated or<br />

with specific TLR ligands. After culture the cells were<br />

stained for flow cytometric analysis with intracellular<br />

anti-cytokine antibodies.<br />

Results: TLR2 levels increased within four weeks of<br />

effective therapy in HBeAg-positive patients (P


level of TLR4 expression did not differ significantly<br />

between the groups. The functional relevance of these<br />

findings was established by the demonstration in<br />

HBeAg positive patients of initial reduced cytokine<br />

production (TNF and IL-6) and phospho-p38<br />

kinase production after stimulation of monocytes<br />

with specific TLR ligands. This immunosuppresion<br />

improved rapidly during treatment (P


Abstract 48<br />

Therapeutic Vaccination Against<br />

Chronic Hepatitis C Virus<br />

Infection: Towards a Proof-ofconcept<br />

in Man?<br />

CS Klade 1 , H Wedemeyer 2 , C Sarrazin 3 , E Schuller 1 ,<br />

K Lingnau 1 , MP Manns 2 , and E Tauber 1<br />

1 Intercell AG, Campus Vienna Biocenter, 1030 Vienna,<br />

Austria; 2 Hannover Medical School, Center for Internal<br />

Medicine, Carl-Neuberg-Str. 1, 30625 Hannover,<br />

Germany; 3 Saarland University Hospital, Internal<br />

Medicine II, Kirrberger Straße, 66424 Homburg/Saar,<br />

Germany<br />

Novel approaches for treatment of chronic HCV<br />

infection including therapeutic vaccination are<br />

urgently needed. We have comprehensively identified<br />

disease relevant epitopes applying PBMC from<br />

therapy responders and spontaneous resolvers. IC41<br />

is a prototypic peptide vaccine containing CD8 and<br />

CD4 T cell epitopes and poly-L-arginine as synthetic<br />

adjuvant. Its safety, immunogenicity and occasional<br />

RNA responses in patients refractory to standard<br />

therapy (ST) have been reported. Correlation of<br />

immune and RNA response showed that CD4<br />

proliferation or IFN-gamma ELIspot were not<br />

sufficient for RNA response, but a prerequisite for<br />

IFN-gamma CD8 T cells. Tetramer specific CD8 T cells<br />

showed a partial shift from CCR7+ central memory<br />

to CCR7- effector memory phenotype, also not<br />

associated with RNA decline. The single parameter<br />

correlating was IFN-gamma CD8 CTL above a critical<br />

threshold. Responses were dominated by NS3-1073,<br />

and in one patient an amino acid exchange resulting<br />

in reduced epitope recognition emerged prior RNA<br />

rebound. This evidence for mutational T cell epitope<br />

escape corroborates a causal relationship of T cell<br />

induction and HCV RNA decline. Next, IC41 was<br />

investigated as late add-on to ST: 35 genotype 1<br />

patients, RNA-negative (


Abstract 49<br />

Amelioration of Metabolic<br />

Disturbances and Oxidative Stress<br />

in Hepatitis C Viral Infection by<br />

FK506 (Tacrolimus)<br />

K Moriya 1 , H Miyoshi 1 , S Shinzawa 1 , T Tsutsumi 1 ,<br />

H Fujie 1 , Y Shintani 1 , H Yotsuyanagi 1 , T Suzuki 2 ,<br />

T Miyamura 2 , Y. Matsuura 3 , and K Koike 1<br />

1 University of Tokyo, Tokyo, Japan; 2 National Institute<br />

of Infectious Diseases, Tokyo, Japan; 3 Osaka University,<br />

Osaka, Japan<br />

BACKGROUND: Oxidative stress is assumed to play<br />

a pivotal role in the pathogenesis of liver diseases<br />

in hepatitis C virus (HCV) infection, and the<br />

mitochondria dysfunction may be responsible for<br />

it. FK506 (Tacrolimus) is supposed to protect the<br />

mitochondrial respiratory function. We determined<br />

whether or not FK506 protects the function of<br />

mitochondria and contributes to improvement of<br />

HCV-associated liver disease.<br />

METHODS: FK506 was administered to HepG2<br />

cells expressing HCV core protein or HCV core gene<br />

transgenic mice, which develop hepatic steatosis,<br />

insulin resistance and liver cancer.<br />

RESULTS: FK506 reduced the production of oxidative<br />

stress, dose-dependently and significantly, in coregene-expressing<br />

cells compared to the control cells.<br />

There was a significant reduction in lipid content and<br />

the concentration of carbon 18 monounsaturated<br />

fatty acids by FK506 in core-gene-expressing cells.<br />

In addition, administration of FK506 thrice a week<br />

for three months to HCV core gene transgenic mice<br />

resulted in a significant improvement of hepatic<br />

steatosis. Furthermore, FK506 treatment significantly<br />

lowered the serum insulin level that is provoked by the<br />

core protein, in addition to the reduction of oxidative<br />

stress and DNA damage in core gene transgenic mice.<br />

Taken together, our results indicate the antioxidant<br />

nature of FK506, which blocks oxidative stress<br />

production in hepatocytes expressing the core protein<br />

both in vitro and in vivo.<br />

CONCLUSION: FK506 reversed the effect of the core<br />

protein in the pathogenesis of HCV infection. This<br />

result may provide a new therapeutic aid for chronic<br />

hepatitis C, in which oxidative stress and metabolic<br />

abnormalities in lipid and glucose contribute to liver<br />

pathogenesis.<br />

Abstract 50<br />

Poor Costimulatory Effects<br />

of CD 137 in Patients with<br />

Hepatocellular Carcinoma<br />

JW Shin 1 , SJ Park 2 , NH Park 1 , and YJ Lee 2<br />

1 Department of Internal Medicine, University of Ulsan<br />

College of Medicine, Ulsan University Hospital, Ulsan,<br />

Korea; 2 Department of Internal Medicine, College of<br />

Medicine, Inje University, Busan Paik Hospital, Busan,<br />

Korea<br />

Background/Aims: The 4-1BB, a member of TNF<br />

receptor superfamily, is expressed on activated T<br />

cells and antigen presenting cells. 4-1BB generates<br />

costimulatory signals which involved T cell activation<br />

and proliferation, and tumor suppression. s4-<br />

1BB which is generated by proteolytic cleavage or<br />

alternative splicing inhibits biological activities of<br />

4-1BB. High circulating levels of s4-1BB have been<br />

suggested to suppress immune response and this<br />

finding was observed in hematologic malignancy.<br />

In this study, we investigated expression of 4-1BB<br />

on peripheral blood mononuclear cell (PBMC) and<br />

serum concentration of s4-1BB in patients with<br />

Hepatocellular carcinoma (HCC) and healthy control.<br />

We also analyzed correlations between s4-1BB<br />

concentrations and clinical characteristics of HCC<br />

patients.<br />

Methods: 4-1BB expressions on PBMC from twenty<br />

three HCC patients and twenty four healthy controls<br />

were analyzed by flow cytometry after stimulation<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 53


with CD3. Serum levels of s4-1BB were measure by<br />

an enzyme linked immunosorbent assay.<br />

Results: The expression of 4-1BB was significantly<br />

lower on PBMC of HCC patients than that of healthy<br />

control (10.35 ± 5.3 % vs 23.08 ± 6.73%; p


is currently under phase III clinical trial in a larger<br />

number of chronic hepatitis B patients.<br />

Abstract 52<br />

HBsAg-HBIg Complex Modulates<br />

Antigen Presentation of Dendritic<br />

Cells from Chronic Hepatitis B<br />

Patients<br />

highest in DCs stimulated with YIC. When T cells<br />

from patients were incubated with YIC-loaded DCs,<br />

higher numbers of cells produced IFN-γ, IL-2 and L-5,<br />

IL-10 were observed.<br />

CONCLUSIONS: YIC surpasses HBsAg or anti-HBs<br />

in modulating DC functions from chronic hepatitis B<br />

patients in vitro.<br />

B Zheng 1 , X Yao 2 , J Zhou 1 , and Y-M Wen 2<br />

1 Department of Microbiology, the Hong Kong University,<br />

Hong Kong SAR, China; 2 Key Laboratory of Medical<br />

Molecular Virology, Shanghai Medical College, Fudan<br />

University, Shanghai, China<br />

BACKGROUND: YIC (HBsAg complexed with anti-<br />

HBs) has shown to revert immune tolerance in a<br />

transgenic mouse model. In phase I and II clinical<br />

trials, YIC induced high titer of anti-HBs in healthy<br />

adults, and serum IFN-γ and IL-2 in chronic hepatitis<br />

B patients. The highest rates of HBeAg loss (23.1%),<br />

HBeAg sero-conversion (21.8%), and higher than<br />

2 log 10<br />

decrease in serum HBV DNA (31.2%) were<br />

observed in the 60 µg YIC immunized group of chronic<br />

hepatitis B patients. The therapeutic mechanism of<br />

YIC was predicted to modulate antigen presenting cells<br />

by the Fc fragments of anti-HBs in YIC. This study is<br />

thus to investigate how YIC modulates dendritic cell<br />

(DCs) functions from chronic hepatitis B patients.<br />

METHODS: After DCs from chronic hepatitis B<br />

patients were separately incubated with yeastderived<br />

HBsAg, HBsIg or YIC, HLA II, CD80, CD86,<br />

CD40 molecules on DCs were monitored by FACS,<br />

and IL-12 levels were monitored using ELISA kits.<br />

T cells from the same patient were incubated with<br />

their DCs previously loaded with HBsAg, HBIg or<br />

YIC and numbers of Th cells produced cytokines were<br />

determined by ELISPOT.<br />

RESULTS: The mean HLA II, CD 86 and CD40<br />

molecules per dendritic cell were the highest in<br />

samples incubated with YIC. IL-12 was also the<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 55


New Therapeutic<br />

Approaches<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 57


ABSTRACT 53<br />

HCV Entry Pathways and<br />

Implications for the Development<br />

of Entry Inhibitors<br />

MJ Evans, T von Hahn, AJ Syder, A Ploss,<br />

DM Tscherne, and CM Rice<br />

Center for the Study of Hepatitis C, The Rockefeller<br />

University, New York, USA<br />

The hepatitis C virus (HCV) is a serious global<br />

public health problem, yet many aspects of the viral<br />

replication cycle remain mysterious. In particular,<br />

only basic steps required for viral entry into a host cell<br />

have been defined. Historically, technical limitations<br />

have made this a difficult step in the HCV replication<br />

cycle to study. However, recently developed systems<br />

including retroviral pseudotyped particles harboring<br />

functional HCV glycoproteins (HCVpp) and cell culture<br />

produced infectious particles (HCVcc) now enable<br />

the study of HCV infection of cultured cells. While<br />

HCVcc provides a means to study the entry processes<br />

of authentic HCV virions, HCVpp appear to mimic all<br />

early entry processes of HCV. Furthermore, HCVpp<br />

can infect more divergent cell types that may lack<br />

the factors necessary for efficient RNA replication of<br />

HCVcc. Through the use of these systems, HCV entry<br />

has been defined as a temperature and pH-dependent,<br />

clathrin-mediated process requiring at least several<br />

cellular molecules, including the tetraspanin CD81,<br />

scavenger receptor class B type I (SR-BI), and perhaps<br />

glycosaminoglycans (GAGs). We have recently<br />

identified claudin-1 (CLDN1), a component of tight<br />

junction complexes, as an essential HCV entry<br />

factor. Even this list of HCV entry factors appears<br />

incomplete, as numerous cell lines express all of them<br />

yet remain uninfectable.<br />

Despite great progress on developing specific HCV<br />

antivirals, it is clear that the emergence of viral<br />

resistance will be a problem facing future HCV<br />

treatment options. Thus it is a priority for HCV<br />

research to develop a spectrum of inhibitors targeting<br />

diverse steps in the virus lifecycle. HCV cell entry may<br />

represent one such unique stage of the HCV replication<br />

cycle that can be targeted. Without a complete picture<br />

of the dynamics of HCV infection within the host, it<br />

is difficult to speculate how effective such inhibitors<br />

would be in a therapeutic setting. HCV cell entry<br />

inhibitors may be exceptionally useful post-liver<br />

transplantation, where universal graft reinfection<br />

frequently results in rapid fibrosis progression and<br />

subsequent graft failure. A greater understanding of<br />

the HCV entry process, including the definition of the<br />

complete set of entry factors and their specific roles,<br />

may be required for the development of therapies<br />

targeting HCV entry. In addition, the identification<br />

of the tight junction protein CLDN1 as an essential<br />

HCV entry factor suggests the importance of<br />

understanding HCV entry into polarized cells, which<br />

may greatly impact the HCV entry process. Such<br />

studies may lead to an understanding of why HCV<br />

selectively infects human hepatocytes with obvious<br />

implications for developing transgenic mouse models<br />

that support productive HCV infection.<br />

Abstract 54<br />

The Role of CD81 and Scavenger<br />

Receptor Class B Member I in HCV<br />

Entry<br />

J Timpe, HJ Harris, J Grove, C Mee, P Balfe, and<br />

JA McKeating<br />

Hepatitis C Research Group, Division of Immunity and<br />

Infection, University of Birmingham, Vincent Drive,<br />

Birmingham, West Midlands, UK, B15 2TT<br />

The selective association of a virus with a target<br />

cell is initially defined by interactions between the<br />

viral encoded glycoproteins and specific cell surface<br />

molecules or viral receptors. The observation that<br />

HCVpp show a restricted tropism for human liver<br />

cells suggests that liver-specific receptors may help<br />

define HCV tropism. Recent evidence suggests the<br />

involvement of at least three host cell molecules in<br />

HCV entry: the tetraspanin CD81, scavenger receptor<br />

class B member I (SR-BI) and the tight junction<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 59


protein Claudin-1 (CLDN1). Other factors, such as<br />

glycosaminoglycans and low density lipoprotein<br />

receptor, have also been implicated in HCV entry,<br />

although their role is less well established.<br />

Since the primary reports describing soluble<br />

truncated HCV E2 interacting with CD81 and SR-BI,<br />

investigators have sought to investigate the role of<br />

these molecules in the viral entry process. Antibodies<br />

specific for CD81 and recombinant forms of the<br />

second extracellular loop of CD81 (sCD81) inhibit<br />

the infectivity of cell bound particles, suggesting that<br />

CD81 does not confer viral attachment and acts as a coreceptor<br />

mediating virus internalization. In contrast,<br />

SR-BI appears to define the primary attachment of<br />

HCV to target cells. Over-expression of SR-BI in Huh-<br />

7.5 hepatoma cells promotes HCV infection, leading<br />

to an increase in focal size and suggesting that SR-BI<br />

facilitates cell-cell transmission of infectivity.<br />

To quantify cell-cell transfer of HCV infectivity we<br />

developed an infectious center assay, where infected<br />

cells are co-cultured with fluorescently labelled naïve<br />

target cells in the presence or absence of neutralizing<br />

antibodies to inhibit the infectivity of cell-free<br />

virus. Enumeration by flow cytometry and indirect<br />

immunofluorescence demonstrated efficient cell-cell<br />

transmission of HCV infectivity. sCD81 and anti-<br />

CD81 inhibit cell-free particle infection of Huh-7.5<br />

and partially reduce cell-cell transmission. CD81<br />

negative HepG2 hepatoma cells, which are resistant<br />

to cell-free virus infection, became infected after coculturing<br />

with JFH-1 infected cells, confirming that<br />

CD81 independent routes of cell-cell transmission<br />

exist. Target cell expression of SR-BI and CLDN1<br />

promotes cell-cell transfer of infectivity. These data<br />

demonstrate that HCV can transmit in vitro by cellfree<br />

virus infection and by direct transfer between<br />

cells and these different routes of transmission may<br />

utilize different receptors.<br />

Imaging techniques that take advantage of fluorescence<br />

resonance energy transfer between fluorescent<br />

tagged receptors allow us to study localization<br />

and protein associations in non-polarized and<br />

polarized cell systems. These data allow us to model<br />

the stoichiometry of the receptor molecules and<br />

provide information on novel targets for antiviral<br />

therapy.<br />

Abstract 55<br />

Lipid Droplet is an Important<br />

Organelle for Production of<br />

Infectious Hepatitis C Virus<br />

K Shimotohno<br />

Center for Integrated Medical Research, Keio University,<br />

Tokyo, Japan<br />

BACKGROUND: Hepatitis C virus (HCV) is a<br />

causative agent of chronic hepatitis, cirrhosis and<br />

hepatocellular carcinoma. Patients infected with HCV<br />

often associate with diseases such as diabetes as well as<br />

steatosis, which are believed to be promoting factors<br />

to the development of hepatocellular carcinoma.<br />

Animal model experiments showed that HCV capsid<br />

protein (Core) was involved in the development of<br />

these diseases. HCV Core regulates metabolism of<br />

lipid synthesis, which may result in accumulation of<br />

lipid in cells. Moreover, HCV Core often associates<br />

with the lipid droplet (LD) in cells expressing Core<br />

solitary. However, the role of lipid accumulation as<br />

well as Core-association to the LD on HCV replication<br />

remains elusive.<br />

METHOD: Wild and mutated HCV replicons derived<br />

from JFH1, an infectious molecular clone of HCV-<br />

2a, were introduced into HuH7 cells and subcellular<br />

localization of HCV proteins was analyzed in<br />

connection with production of infectious virus into<br />

culture medium.<br />

RESULT: HCV Core protein associated with the LD,<br />

which not only confirmed the previous reports from<br />

other group but also showed the association of Core<br />

even when expressed with whole HCV proteins. In<br />

addition, we observed close association of other virus<br />

proteins with the LD in Core-dependent manner. This<br />

observation was further confirmed by the following<br />

60 Global Antiviral Journal Volume 3, Supplement 2


evidence; (1) Core which lacks association with the<br />

LD failed to recruit other HCV proteins to the LD, (2)<br />

No association of other HCV proteins with the LD was<br />

observed in HuH7 cells bearing HCV replicon lacking<br />

Core production. Infectious HCV was produced only<br />

in those cells expressing HCV proteins that associate<br />

with the LD. Importanly, non-infectious HCV was<br />

released from cells irrespective to association of HCV<br />

proteins with the LD.<br />

DISCUSSION: Lipid droplet plays important roles<br />

in production of infectious HCV particle. Our result<br />

suggests that Core plays major roles to recruit other<br />

HCV proteins around the LD, which may generate the<br />

environment in where production of infectious virion<br />

proceeds. Accumulation of lipid in cells upon HCV<br />

infection may be requisite for efficient production of<br />

HCV progeny and also may play as a trigger to develop<br />

steatosis.<br />

Abstract 56<br />

The Role of Cyclophilins and<br />

Cyclophilin Inhibitors in the<br />

Replication of HCV<br />

R Crabbé<br />

Debiopharm, Switzerland<br />

Cyclophilins are ubiquitously present proteins with<br />

peptidyl-prolyl cis-trans isomerase activity that<br />

play an important role in protein folding and in<br />

isomerization of native proteins in several cellular<br />

systems. Cyclophilin B (CypB) is targeted to the<br />

secretory pathway via an endoplasmic reticulum<br />

signal sequence. It is involved in the regulation of<br />

inflammatory processes, mainly through interaction<br />

with CD147 and is also a potent chemotactic agent. It<br />

has recently been suggested that it plays a role in HCV<br />

replication. Growing evidence indicates that CypB is a<br />

positive modulator of the HCV RNA dependent RNA<br />

polymerase in the replication complex. CypB may act<br />

as a functional regulator of NS5B RNA-dependent<br />

RNA polymerase and cyclophilin inhibitors seem<br />

to interfere with this interaction. Consistent with<br />

this finding, a model for the role of CypB in HCV<br />

replication has been proposed. CypB interaction<br />

with NS5B enhances RNA binding and promotes<br />

RNA replication. Cyclosporin A and other cyclophilin<br />

inhibitors would therefore block the ability of CypB to<br />

interact with NS5B, giving rise to weaker RNA binding<br />

and the inability to form a functional RNA replication<br />

complex. A reduced HCV replication of replicon cell<br />

lines by shRNA after knockdown of CypA, CypB, or<br />

CypC has also been reported. Therefore, the precise<br />

mechanism of interaction of CypB and/or other Cyps<br />

with the RNA-dependent RNA polymerase NS5B or<br />

other HCV proteins still needs to be elucidated. It<br />

remains unknown if all HCV genotypes exploit Cyp(s)<br />

in the same manner.<br />

Recently, CsA derivatives lacking immunosuppressive<br />

action have been synthesised. These drugs (NIM811,<br />

Debio 025 and SCY-635) showed potent anti-HCV<br />

activity in the replicon system with IC 50<br />

values of 0.07<br />

μM to 0.22 μM for Debio 025 and from 0.35 to 0.66 μM<br />

for NIM811. Debio 025, alone or in combination with<br />

other anti-HCV drugs, was also particularly efficient<br />

in curing replicon containing cells from their replicon,<br />

while Cyclosporin A or a HCV protease inhibitor<br />

did not result in clearance of the HCV replicon. The<br />

only cyclophilin inhibitor for which patient data are<br />

available is Debio 025. In HIV-1 and HCV co-infected<br />

subjects with compensated liver disease, a dose of<br />

1200 mg BID of Debio 025 for 14 days resulted in a<br />

3.6 log 10<br />

decrease of viral load.<br />

The present data indicate that cyclophilins seem to play<br />

an important role in the replication of the hepatitis C<br />

virus. Although the exact mechanism of action at the<br />

molecular level is not yet fully elucidated, Cyclophilin<br />

inhibition seems to represent a new approach to anti-<br />

HCV treatment mainly by interfering at the level of<br />

the host-viral interaction.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 61


Abstract 57<br />

HCV Infection Increases Claudin-1<br />

and Claudin-7 Expression by<br />

Inducing Cirrhosis<br />

A Kiss 1 , A Holczbauer 1 , E Batmunkh 1 , G Lotz 1 ,<br />

P Kupcsulik 2 , and Z Schaff 1<br />

1 Semmelweis Medical University, 2nd Inst. of Pathology,<br />

Budapest, Hungary; 2 Semmelweis Medical University, 1st<br />

Inst. of Surgery, Budapest, Hungary<br />

BACKGROUND: Claudins (CLDNs) (1-24) have been<br />

recently identified as integral proteins of tight junction<br />

strands. They have been reported to be differentially<br />

regulated in malignancies and implicated in the<br />

process of carcinogenesis and tumor progression.<br />

Changes in claudin-1 and claudin-10 expression have<br />

been recently found in the development of primary<br />

hepatocellular carcinoma (HCC). Three host cell<br />

molecules have been reported as significant entry<br />

factors for hepatitis C virus (HCV): beside CD81 and<br />

scavenger receptor B I. claudin-1 has been recently<br />

described as co-factor in the entry of HCV into<br />

hepatocytes. Further, claudin-6 and claudin-9 may<br />

function as additional co-receptors for HCV as well.<br />

The objective was to characterize the mRNA and<br />

protein expression of claudin-1, 2, 3, 4 and 7 in HCCs,<br />

surrounding and normal livers with respect to HCV<br />

infection and the presence of cirrhosis.<br />

between the presence of cirrhosis and elevated<br />

levels of CLDN-1 and -7 protein expression while<br />

no correlation was revealed between HCV infection<br />

and claudin expression. Claudin-1 and 7 protein<br />

expression was indeed significantly elevated in<br />

cirrhosis (2.86-9.25 fold) compared with normal liver<br />

and non-cirrhotic surrounding liver. HCC developed<br />

in cirrhotic livers showed even higher expression of<br />

claudin-1 contrary to decreased CLDN-7 expression<br />

in HCC when compared to cirrhosis. Western blot<br />

analysis confirmed immunohistochemical data.<br />

mRNA expression of claudin-1 and -7 regarding to<br />

the presence of cirrhosis showed similar differences<br />

as found in protein expression.<br />

CONCLUSIONS: Our data indicate that cirrhosis<br />

finally increases the expression of claudin-1 and -7<br />

in the surrounding liver and in the developing HCCs<br />

as well. Since HCV infection alone does not alter<br />

claudin expression dramatically our data suggest<br />

that the elevated claudin-1 and -7 expression is the<br />

result of an indirect effect of HCV infection achieved<br />

by the induction of cirrhosis. This would be a new<br />

mechanism how HCV virus infection and following<br />

hepatitis and cirrhosis could enhance the effectivity<br />

of viral entrance and therefore boost the chronicity<br />

of the viral infection. The project was supported<br />

by grants: OTKA-T049559, ETT-049/2006, NKFP<br />

1A/002, ETT-156/2006, NKFP 0056/2004<br />

METHODS: 25 surgically resected HCCs with surrounding<br />

tissues and ten normal livers were examined<br />

by real-time RT-PCR and immunohistochemistry<br />

and Western blot analysis. Immunoreactivity of<br />

CLDNs were quantified by morphometry.<br />

RESULTS: Claudin-1 and -7 protein expression was<br />

significantly elevated in HCV infected surrounding<br />

livers and HCCs when compared to normal liver<br />

samples. On the other hand HCCs or surrounding<br />

livers of HCV infected samples did not show significant<br />

alteration in claudin 1, 2, 3, 4 and 7 mRNA or protein<br />

expression compared to HCV negative specimens.<br />

However, Spearman analysis indicated correlation<br />

62 Global Antiviral Journal Volume 3, Supplement 2


Abstract 58<br />

Development of Novel<br />

Hyperglycosylated Type 1<br />

Interferons: A Strategy to Improve<br />

PK Performance Without Loss of<br />

Biological Potency<br />

LM Blatt<br />

Alios BioPharma Inc., South San Francisco, California,<br />

USA<br />

Interferons are cytokines that are induced as a<br />

consequence of viral and microbial infections that<br />

have pleiotropic biological effects that play a role<br />

in modulating innate and adaptive immunity. Over<br />

the past decade, the use of type 1 interferon to treat<br />

chronic hepatitis C has led to sustained virologic<br />

response in approximately 50% of patients. Although<br />

the use of interferons has become widespread,<br />

several challenges still exist with respect to treatment<br />

optimization and at least half of the patients cannot<br />

obtain a sustained response to existing therapies.<br />

These challenges include poor PK profiles, limited<br />

biological activity, and suboptimal therapeutic<br />

indices. The addition of polyethylene glycol to type<br />

1 interferon has been shown to dramatically increase<br />

plasma exposure following dosing and has led to<br />

increased response rates in the treatment of chronic<br />

hepatitis C. It is important to note that the addition<br />

of polyethylene glycol to recombinant proteins has<br />

the effect of reducing the specific activity of the<br />

molecule in vitro. This is due to the fact that the large<br />

polyethylene glycol molecule that is necessary to<br />

block renal filtration can sterically hinder the active<br />

binding of the interferon to the cell surface receptor.<br />

were identified that were quantitatively modified by<br />

addition of carbohydrate moieties. A single variant<br />

carrying both sites was quantitatively modified at<br />

both sites. Glycosylation substantially increased<br />

molecular weight with no resultant loss of biological<br />

potency. In cell culture models, consensus interferon<br />

demonstrates increased potency when compared to<br />

naturally occurring type 1 interferons. Comparisons<br />

of the antiviral potency of glycol-variants with the<br />

parent non-glycosylated consensus interferon and<br />

PEG-IFN alfa 2a using the VSV on A549 cells are<br />

shown in the table below:<br />

IFN<br />

EC 50<br />

(pg/mL)<br />

Consensus IFN 1.83 ± 0.41<br />

Single Glyco-A 2.53 ± 0.53<br />

Single Glyco-B 3.98 ± 0.85<br />

Double Glyco-A+B 2.27± 0.41<br />

PEG-IFN alfa 2a 1200± 600<br />

Statistical analysis of the EC 50<br />

values obtained<br />

demonstrated that all glycol-variants retain similar<br />

biological potency when compared to the consensus<br />

interferon molecule and all are approximately 1000X<br />

more potent when compared to PEG-IFN alfa 2a<br />

(P


Abstract 59<br />

Bioinformatics Resources<br />

Supporting the Analysis of<br />

Hepatitis C Virus<br />

EJ Lefkowitz 1 , C Kuiken 2 , B Peters 3 , A Sette 3 , and<br />

C Upton 4<br />

1 University of Alabama at Birmingham, Birmingham, AL,<br />

USA; 2 Los Alamos National Laboratory, Los Alamos, NM,<br />

USA; 3 The La Jolla Institute for Allergy and Immunology,<br />

La Jolla, CA, USA; 4 University of Victoria, Victoria, BC,<br />

Canada<br />

Several groups that provide bioinformatics resources<br />

to the scientific community to aid in the study<br />

of Hepatitis C virus (HCV) have recently begun<br />

collaborating to expand the available informational<br />

and analytical tools supporting HCV research. The<br />

Viral Bioinformatics Resource Center (VBRC), the<br />

Immune Epitope Database and Analysis Resource<br />

(IEDB), and the Hepatitis C virus resource at the<br />

Los Alamos National Laboratories (HCV-LANL) are<br />

coordinating the acquisition of new and existing data,<br />

the annotation of that data, and the development<br />

of new analytical tools, to maximize efficiency and<br />

provide cross-connected web sites to access to all of<br />

these resources.<br />

The VBRC (www.vbrc.org) is one of eight NIHsponsored<br />

Bioinformatics Resource Centers established<br />

to make available informational and<br />

analytical resources to the scientific community. The<br />

goal of these Centers is to aid research directed at<br />

providing a better understanding of microorganisms<br />

included on the NIH list of priority pathogens.<br />

The VBRC was specifically directed to study<br />

viruses belonging to the Arenaviridae, Bunyaviridae,<br />

Filoviridae, Flaviviridae, Paramyxoviridae, Poxviridae,<br />

and Togaviridae families, and includes HCV. In addition<br />

to sequence data, the VBRC provides curation for<br />

each of the viral genomes and gene records, resulting<br />

in a searchable, comprehensive mini-review of gene<br />

function relating genotype to biological phenotypewith<br />

special emphasis on pathogenesis. The VBRC<br />

also provides a variety of analytical tools on its web<br />

site to aid in the understanding of the available data,<br />

including tools for genome annotation, comparative<br />

analysis, whole genome alignments, and phylogenetic<br />

analysis.<br />

The IEDB (www.immuneepitope.org) project catalogs<br />

and organizes information regarding antibody and T<br />

cell epitopes from infectious pathogens, experimental<br />

antigens, and self-antigens, and now includes HCV<br />

epitopes. The IEDB contains information on epitopes<br />

curated manually from the scientific literature. Both<br />

intrinsic structural and phylogenetic features, as well<br />

as information relating to the interactions of the<br />

epitopes with the host’s immune system are stored,<br />

and a variety of tools for querying the database and<br />

analyzing epitope information are provided.<br />

The HCV-LANL resource (hcv.lanl.gov) provides<br />

access to a database of annotated HCV sequences<br />

that includes detailed isolate and host (patient)<br />

information. The sequence diversity of HCV requires<br />

methods to track variants and assess their association<br />

with individual infections and with epidemiologically<br />

related outbreaks. This interactive resource provides<br />

flexible retrieval tools for sequences, clinical<br />

information, and meta-data, as well as utilities for<br />

scientific data analysis, including tools to assist in the<br />

analysis of sequence variation.<br />

By distributing responsibilities, sharing data, and<br />

interacting with the scientific community, these three<br />

groups will expand existing resources to establish an<br />

even more comprehensive set of easy-to-use data<br />

query and analytical tools that provide a useful and<br />

used platform supporting HCV research.<br />

64 Global Antiviral Journal Volume 3, Supplement 2


Abstract 60<br />

A Combination of Direct Antiviral<br />

Compounds Can Achieve<br />

Sustained Viral Response in<br />

Hepatitis C Virus-infected<br />

Chimpanzees<br />

DB Olsen 1 , L Handt 1 , K Koeplinger 1 , S Ludmerer 1 ,<br />

D Graham 1 , M MacCoss 2 , NJ Liverton 1 , JP Vacca 1 ,<br />

JA McCauley 1 , D Hazuda 1 , and SS Carroll 1<br />

1<br />

Merck Research Laboratories, West Point, PA, 19486,<br />

USA; 2 Merck Research Laboratories, Rahway, NJ, 07065,<br />

USA<br />

BACKGROUND: Current pegylated interferon-α and<br />

ribavirin combination therapies to treat infection by<br />

hepatitis C virus (HCV) have significant side effects<br />

and show limited efficacy in patients infected with<br />

HCV genotype 1. Efforts to develop novel therapies<br />

that enhance both efficacy and tolerability have<br />

focused on direct antiviral agents targeting the virally<br />

encoded RNA polymerase, NS5B, and protease,<br />

NS3/4A.<br />

METHODS: To explore the potential to achieve<br />

sustained virologic response by administration<br />

of a combination of polymerase and protease<br />

inhibitors, HCV-infected chimpanzees were dosed<br />

with MK-0608, a nucleoside analog inhibitor of<br />

HCV RNA polymerase, or with a novel, macrocyclic<br />

inhibitor of NS3/4A protease, or a combination of<br />

both compounds. Plasma samples were collected<br />

at time points before, during and after the dosing<br />

period and plasma viral loads determined using the<br />

Taqman assay (limit of quantitation, LOQ, 20 IU/<br />

mL). Population sequencing and a novel allele specific<br />

Taqman assay (AUGER) were used to assess levels of<br />

viral variants known to be resistant to inhibition by<br />

the two compounds.<br />

RESULTS: Administration of potent inhibitors of<br />

either NS5B or NS3/4A to HCV-infected chimpanzees<br />

results in profound suppression of viral loads in<br />

plasma. All of the chimpanzees receiving combination<br />

dosing experienced decreases in plasma viral titer<br />

to below the LOQ. The viral titer of one of three<br />

chimpanzees receiving the longest duration of<br />

combination dosing remained below the LOQ 26<br />

weeks after the end of dosing. Viral variants resistant<br />

to inhibition by the NS3/4A inhibitor were detected<br />

in rebounding viral populations in the other animals.<br />

CONCLUSIONS: The results demonstrate that<br />

profound long-term suppression of viral titer can<br />

be achieved by administration of a combination of<br />

direct antiviral agents in the chimpanzee model of<br />

HCV infection. The results of these studies regarding<br />

the efficacy, duration of response, and potential for<br />

development of antiviral resistance have implications<br />

for designing strategies to achieve SVR with direct<br />

antiviral therapies in HCV infected patients.<br />

Abstract 61<br />

Potent Antiviral Activity of the<br />

Nucleoside HCV Inhibitor, R7128,<br />

in Prior IFN Non-responders<br />

JG McHutchison 1 , R Reddy 2 , M Rodriguez-Torres 3 ,<br />

E Gane 4 , R Robson 5 , J Lalezari 6 , GT Everson 7 ,<br />

E DeJesus 8 , HE Vargas 9 , A Beard 10 , GZ Hill 11 , M Otto 10 ,<br />

W Symonds 10 , and M Berrey 10<br />

1 Duke Clinical Research Institute, Durham, NC, USA;<br />

2 University of Pennsylvania, Philadelphia, PA, USA;<br />

3 Fundacion de Investigacion d Diego, Santurce, PR,<br />

USA; 4 Auckland Clinical Studies Limited, Auckland, New<br />

Zealand; 5 CCST, Christchurch, New Zealand; 6 Quest<br />

Clinical Research, San Francisco, CA, USA; 7 University<br />

of Colorado, Aurora, CA, USA; 8 Orlando Immunology<br />

Center, Orlando, FL, USA; 9 Mayo Clinic, Phoenix, AZ,<br />

USA; 10 Pharmasset, Inc., Durham, NC, USA; 11 Roche,<br />

Palo Alto, CA, USA<br />

Background: R7128 is a pro-drug of PSI-6130, a<br />

cytidine nucleoside analog polymerase inhibitor, for<br />

treatment of HCV. Safety and PK have been evaluated<br />

following single oral doses in healthy volunteers;<br />

preliminary antiviral activity was assessed following<br />

14d R7128 in subjects with HCV genotype 1<br />

infection.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 65


Methods: Single doses of R7128 were administered<br />

to 46 healthy subjects. Six active and two placebo<br />

subjects per group were enrolled in 5 sequential dose<br />

groups (500 mg, 1500 mg, 4500 mg, 6000 mg, and<br />

9000 mg) and a food effect group (1500 mg). In Part<br />

2, multiple oral doses of R7128 were administered<br />

for 14 days to 40 HCV-infected patients (8 active &<br />

2 placebo per cohort) at doses of 750mg QD, 1500mg<br />

QD, 750mg BID & 1500mg BID.<br />

Results: Following single doses, nineteen (19)<br />

adverse events were reported; all were mild to<br />

moderate, none were dose-dependent, and no<br />

gastrointestinal AEs were observed. In the multiple<br />

dose study, all subjects had HCV genotype 1 (30 – 1a;<br />

10- 1b), had previously failed alpha-interferon and<br />

were non-cirrhotic. There were no SAEs reported<br />

and no AEs required dose modification. No clinically<br />

significant changes in vital signs, ECGs, hematology,<br />

renal or other laboratory parameters occurred.<br />

Preliminary data on AEs reported during treatment<br />

in subjects receiving R7128 include a total of 51<br />

events in 18 of 32 subjects, most of mild intensity.<br />

The most frequently reported AEs for patients<br />

receiving R7128 were headache (13) and dry mouth<br />

(3). 34 adverse events occured in 7 subjects receiving<br />

placebo, with headache (4) and diarrhea (4) most<br />

commonly reported. After both single and multiple<br />

doses, plasma exposure to the prodrug, R7128, was<br />

negligible, while concentrations of the active moiety,<br />

PSI-6130 increased less than proportionally with<br />

dose. PSI-6130 C max<br />

occured 2-3 hours after dosing.<br />

The terminal half-life was ~5h for PSI-6130. Mean<br />

plasma HCV RNA in all 4 dose groups decreased<br />

in a dose-dependent manner with placebo values<br />

remaining at baseline. The mean reduction in HCV<br />

RNA with the 1500 mg BID dose was -2.7 log 10<br />

IU/mL<br />

and ranged from -1.2 to -4.2 log 10<br />

(below the limit of<br />

detection) at Day 15.<br />

Conclusions: This study demonstrated that<br />

R7128, a direct antiviral, can deliver sufficient<br />

antiviral potency via monotherapy to suppress HCV<br />

below the level of detection (


METHODS: 104 patients were randomized to: Dual<br />

1500: R1626 1500 mg bid + PEG-IFNα-2a (n=21);<br />

Dual 3000: 3000 mg bid + PEG-IFNα-2a (n=32);<br />

Triple 1500: 1500 mg bid + PEG-IFNα-2a + RBV<br />

(n=31); SOC (standard of care): PEG-IFNα-2a + RBV<br />

(n=20).<br />

RESULTS: At week 4 HCV RNA was undetectable (


Methods: These double-blind studies randomized<br />

mono-infected subjects with HBeAg+ or HBeAg-<br />

CHB 2:1 to TDF or ADV. Entry criteria included 18-<br />

69 years of age, compensated liver disease, a Knodell<br />

necroinflammatory score≥3, ALT >2xULN (HBeAg+)<br />

or >ULN (HBeAg-), HBV DNA>10 6 c/mL (HBeAg+) or<br />

> 10 5 c/mL (HBeAg-). Biopsies were performed pretreatment<br />

and between Weeks 44 and 48. HBV DNA<br />

was measured using the Roche COBAS TaqMan assay<br />

(LLQ=169c/mL).<br />

Results: HBeAg+: 266 nucleos(t)ide naïve subjects<br />

(176 TDF:90 ADV) were randomized and treated;<br />

mean baseline HBV DNA was 8.72 log 10<br />

c/mL and ALT<br />

was 147 IU/mL. HBeAg-: 375 subjects (250 TDF:125<br />

ADV) both naïve and lamivudine experienced (18%)<br />

were randomized and treated for 48 weeks; mean<br />

baseline HBV DNA was 6.9 log 10<br />

c/mL and ALT was<br />

140 IU/mL. More than 90% of the subjects completed<br />

primary endpoint assessments and overall 1% TDFtreated<br />

subjects discontinued due to an adverse<br />

event. A statistically significantly greater response<br />

was observed for TDF-treated subjects: HBV DNA<br />

99% homology to the 5’-<br />

NCR. (ii) BLAST alignments show no assignment to<br />

any known HCV geno-/subtype. (iii) HCV sequence<br />

stretches (about 82 bp) are contained within repeated<br />

68 Global Antiviral Journal Volume 3, Supplement 2


sections containing several annealing sites for the<br />

primers. (iv) Amplified DNA fragments up to 1288<br />

bp with a 23 bp sequence in between show high<br />

homology to a center sequence stretch of the 5’-NCR<br />

(nt 89 to nt 170) of HCV, i.e., with three substitutions<br />

and a gap.<br />

We further could identify CD34(+) cells, enriched<br />

with these DNA sequence section. In particular, this<br />

marker is expressed on hematopoietic progenitor<br />

cells.<br />

Conclusions: Our working hypothesis: These<br />

particular DNA sequences are part of longer ‘extra<br />

chromosomal’ DNA molecules.<br />

This kind of ‘extra chromosomal’ DNA of various<br />

length could have been generated by incremental<br />

acquisition from these numerous short sequence<br />

stretches in the human genome through the activity<br />

of mobile genetic elements, a common feature of<br />

them is to generate repeats in the target sequences.<br />

This may point at different stages during their<br />

evolution.<br />

The findings point at three directions at least: (i)<br />

large parts of the HCV’s 5’-NCR have reached a DNA<br />

level with yet unknown functioning independently of<br />

those (ii) being part of an infectious RNA unit named<br />

HCV. (iii) These HCV homologous sequences could be<br />

part of non-coding RNA families.<br />

Abstract 65<br />

A Novel Anti-viral Compound,<br />

BIT225, Inhibits Bovine Diarrhea<br />

Virus (BVDV) In Vitro, and has<br />

Synergistic Antiviral Activity in<br />

Combination with Recombinant<br />

Interferon Alpha-2b (rIFNα−2b)<br />

and Ribavirin<br />

CA Luscombe 1 , Z Huang 2 , M Murray 2 , M Miller 1 and<br />

G Ewart 1<br />

1 Biotron Limited, Canberra, ACT, Australia; 2 Hepatitis<br />

Research Program, Southern Research Institute,<br />

Frederick, MD, USA<br />

Background: The hepatitis C virus (HCV) p7<br />

protein is a “viroporin” (virus encoded ion channel),<br />

potentially involved in virus entry and/or assembly<br />

of progeny virions: It is required for HCV infection<br />

in chimpanzees, validating it as a target for antiviral<br />

chemotherapy. Biotron Limited has developed a<br />

library of over 300 potential antiviral compounds,<br />

specifically designed to target viroporins, and has<br />

identified the compound BIT225 as an inhibitor of p7<br />

ion channel activity. BIT225, therefore, has potential<br />

for development as an anti-HCV therapeutic,<br />

particularly as this compound has already successfully<br />

completed phase 1 studies in healthy volunteers with<br />

no significant adverse events reported.<br />

Bovine viral diarrhea virus (BVDV) is commonly<br />

studied as a model system for HCV. BVDV encodes a<br />

homologous p7, ion channel forming protein that is<br />

essential for virus replication.<br />

Methods: A cytoprotection assay was used for<br />

evaluation of compounds against BVDV replication in<br />

Madin-Darby bovine kidney cells in vitro. BIT225 was<br />

tested either alone or in combination with rIFNα-<br />

2b and/or ribavirin. For triple drug combination<br />

experiments, two fixed, sub EC 50<br />

concentrations of<br />

rIFNα-2b (5 and 10 IU/ml) were tested against 8 twofold<br />

dilutions of BIT225 (from 4µM) and 5 two-fold<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 69


dilutions of ribavirin (from 20µg/ml). Effects of the<br />

drug combinations were analysed using MacSynergy<br />

software to determine synergy, additivity or<br />

antagonism.<br />

Results: BIT225 alone inhibits BVDV replication<br />

with an EC 50<br />

value of 314 nM. The EC 50<br />

for rIFNα-2b<br />

alone was 21.7 IU/ml, while ribavirin, which is well<br />

known to enhance the activity of interferons, had<br />

minimal antiviral effect up to 20 µg/ml when tested<br />

alone. In combination with BIT225, ribavirin slightly<br />

antagonized the strong antiviral activity of BIT225,<br />

but only at the high concentrations of both drugs. In<br />

contrast, the combination of rIFNα-2b and BIT225<br />

showed slight to high synergism against BVDV. As an<br />

example, in the presence of 10 IU/ml rIFNα-2b, the<br />

EC 50<br />

value for BIT225 dose response was reduced to<br />

138 nM. The triple drug combinations showed even<br />

higher synergism: Complete virus inhibition was<br />

seen with 31nM BIT225 in the presence of 10 IU/ml<br />

rIFNα-2b and 2.5 µg/ml ribabivirin.<br />

Conclusions: These studies show strong synergism<br />

between BIT225 and interferon. While the<br />

combination of BIT225 and ribavirin is not beneficial<br />

in the absence of interferon, the addition of ribavirin<br />

in a triple compound combination further boosts<br />

antiviral efficacy, allowing all three compounds to<br />

be used at lower concentrations. BIT225 may act by<br />

inhibiting an additional step in the virus replication<br />

cycle (p7 activity) and/or by enhancing the effect<br />

of interferon on the cell. A Phase Ib evaluation of<br />

BIT225 in chronic HCV subjects is planned for the<br />

last quarter of 2007.<br />

Abstract 66<br />

Identification of Therapeutic<br />

Targets in Hepatitis B Virus<br />

(HBV) Associated Hepatocellular<br />

Carcinoma (HCC)<br />

MA Feitelson 1 , Z Lian 1 , J Liu 2 , HY Wang 3 , WC Wu 3 ,<br />

P Arbuthnot 4 , MC Kew 4 , and MM Clayton 1<br />

1 Department of Biology, College of Science and<br />

Technology, Temple University, Philadelphia, PA, USA;<br />

2 Department of Digestive Diseases, Fourth Military<br />

Medical University, Xi’an, P.R. China; 3 Shanghai<br />

Eastern Hospital & Institute of Hepatobiliary Surgery,<br />

Second Military Medical University, Shanghai, P.R.<br />

China; 4 Department of Medicine, University of the<br />

Witwatersrand, Johannesburg, South Africa<br />

BACKGROUND: Intrahepatic expression of hepatitis<br />

B x antigen (HBxAg) is associated with the<br />

development of HCC, perhaps through up-regulated<br />

expression of selected cellular genes.<br />

METHODS: HepG2 cells were stably transduced<br />

with recombinant retrovirus making HBxAg or the<br />

bacterial chloramphenicol acetyltransferase (CAT)<br />

product. RNA isolated from HepG2X and HepG2CAT<br />

cells was subjected to PCR select cDNA subtraction.<br />

Differentially expressed genes from these cultured<br />

cells were validated in clinical samples by in situ<br />

hybridization, northern and western blotting, and<br />

by immunohistochemistry. Selected up-regulated<br />

proteins were then functionally characterized by<br />

individually over-expressing them in liver cell cultures<br />

and then measuring their impact upon hepatocellular<br />

growth, survival and tumor formation. Some of<br />

the underlying signaling molecules regulating cell<br />

growth/survival were also identified.<br />

RESULTS: When expression patterns of cellular<br />

genes were examined in HBxAg positive compared<br />

to negative HepG2 cells, expression of the unique<br />

cellular proteins, up-regulated genes 7 and 11 (URG7<br />

and URG11), were also strongly up-regulated in<br />

liver surrounding HCC and in some tumor nodules.<br />

70 Global Antiviral Journal Volume 3, Supplement 2


URG7 over-expression promoted resistance of liver<br />

cells to Fas and TNFα mediated killing. URG11<br />

over-expression in liver cells strongly promoted<br />

growth in soft agar and accelerated tumorigenesis<br />

in transplantable HepG2 cells. Further examination<br />

showed that over-expression of URG7 and URG11<br />

were associated with up-regulated expression of<br />

wild type β-catenin in liver cell culture, in infected<br />

liver, and in some HBV infected tumors. Extensive<br />

co-staining between HBxAg, URG7, URG11 and<br />

β-catenin was observed in infected liver, suggesting<br />

a close relationship in vivo. Up-regulated expression<br />

of β-catenin correlated with HBxAg trans-activation<br />

function. Transient transfection assays with a<br />

fragment of the β-catenin promoter showed that it was<br />

activated by HBxAg, URG7 and URG11, suggesting<br />

transcriptional activation of the β-catenin gene in<br />

resistance to apoptosis and tumor development.<br />

URG11 specific siRNA partially inhibited the ability<br />

of URG7 to protect against TNFα killing and inhibited<br />

the growth of HCC cells in serum free medium. This<br />

correlated with depressed levels of β-catenin.<br />

CONCLUSIONS: HBxAg, URG7, URG11, and<br />

β-catenin are important therapeutic targets in<br />

hepatocarcinogenesis for the application of existing<br />

drugs and for the development of novel ones.<br />

Abstract 67<br />

A Phase 1, Randomized, Blinded,<br />

Placebo-controlled, Single-dose,<br />

Dose-escalation Study of PEG-<br />

Interferon lambda (PEG-rIL-29) in<br />

Healthy Subjects<br />

DF Hausman 1 , JA Freeman 1 , SM Souza 1 , IA Nestorov 1 ,<br />

and T Zhang 1<br />

1 ZymoGenetics, Inc., Seattle, WA, USA<br />

BACKGROUND: IL-29 is a Type III interferon (IFN)<br />

induced in response to viral infection. IL-29 binds to<br />

a receptor with a more restricted expression pattern<br />

than the IFN-α receptor. PEGylated interferon<br />

lambda or PEGylated recombinant IL-29 (PEG‐rIL-<br />

29) is under development as a potential treatment<br />

for hepatitis C virus (HCV) infection that may be<br />

associated with better tolerability than current IFNs.<br />

PEG-rIL-29 inhibits HCV replication in vitro, and<br />

administration of PEG-rIL-29 to cynomolgus monkeys<br />

results in increased serum beta‐2‐microglobulin<br />

(B2M) and hepatic 2’5’ oligoadenylate synthetase<br />

(OAS). The purpose of this Phase 1 study was to<br />

evaluate the safety, tolerability, pharmacodynamic<br />

and pharmacokinetic profile of a single subcutaneous<br />

dose of PEG-rIL-29 in healthy human subjects.<br />

METHODS: Subjects were randomized 5:1 to receive<br />

PEG‐rIL-29 or placebo on Day 1, with continuous<br />

monitoring for 48 hours post-dose and followup<br />

on Days 4, 8, 15, 29, and 59. Assessments included<br />

standard safety measures, electrocardiogram,<br />

echocardiogram, serum levels of PEG-rIL-29, anti-<br />

PEG‐rIL-29 antibodies, and markers of biological<br />

activity including B2M and OAS.<br />

RESULTS: Seventeen subjects were treated with PEGrIL-29<br />

(5 each at 0.5, 1.5, and 5 μg/kg, and 2 at 7.5<br />

μg/kg) and 3 with placebo. Dose-dependent increases<br />

in B2M were noted starting at 1.5 µg/kg, and in<br />

serum OAS starting at 5 µg/kg. Pharmacokinetics<br />

were dose‐dependent, with the half life estimated<br />

to be 56 hours. PEG-rIL-29 was well tolerated at<br />

doses up to 5µg/kg and was not associated with<br />

fever or significant hematologic or cardiac effects<br />

at any dose level. Dose limiting toxicity, consisting<br />

of reversible Grade 3 alanine aminotransferase<br />

(ALT) elevation associated with Grade 2 increase<br />

in aspartate aminotransferase (AST), occurred in 1<br />

of 2 subjects treated at 7.5 µg/kg. All other adverse<br />

events (AEs) were Grade 1 or 2. The most frequently<br />

reported AEs were pharyngolaryngeal pain (n=3),<br />

increased transaminases/ALT (n=3), leukocytosis<br />

(n=2), contact dermatitis (n=2), and cough (n=2).<br />

Of these, only the transaminase increases and one<br />

event of pharyngolaryngeal pain were considered<br />

related to study drug. Laboratory abnormalities<br />

included dose-dependent Grade 1 decreases in<br />

fibrinogen and transient increases in prothrombin<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 71


time not associated with bleeding or decreases in<br />

platelets. Reversible Grade 1 to 2 elevations in ALT<br />

and AST without associated increases in bilirubin<br />

also occurred in 3/5 subjects treated at 5 µg/kg. No<br />

significant changes were seen in serum chemistries,<br />

renal or hematologic parameters at any dose level. No<br />

subjects developed specific antibodies to PEG-rIL-29.<br />

CONCLUSIONS: Administration of single doses of<br />

PEG-rIL-29 was well tolerated at doses up to 5 µg/kg<br />

without flu-like symptoms or changes in hematologic<br />

parameters and was associated with evidence of<br />

biologic activity. Additional studies are being planned<br />

to further evaluate the safety and potential antiviral<br />

activity of PEG-rIL-29 in subjects with chronic HCV<br />

infection.<br />

Abstract 68<br />

Influence of Laboratory<br />

Parameters on Appearance and<br />

Course of Bleeding in Patients<br />

with Portal Hypertension and<br />

Liver Cirrhosis<br />

L Husova, J Lata, M Senkyrik, and M Dastych<br />

University of Brno, Brno, Czech Republic<br />

BACKGROUND: Acute variceal bleeding is the main<br />

reason of mortality in patients with liver cirrhosis.<br />

The development of esophageal varices as well as<br />

their rupture depends on the level of portal pressure,<br />

however, a number of other factors may play a negative<br />

role in the rise of bleeding and its prognosis.<br />

METHODS: 115 patients with liver cirrhosis were<br />

enrolled (53 with esophageal variceal bleeding, 62<br />

non-bleeding cirrhotic patients. There was a difference<br />

found in Child-Pugh classification (the Childs C<br />

patients were significantly more frequent in the<br />

bleeding group). Immediately after hospitalization; a<br />

panel of hematological (prothrombin time, leukocytes,<br />

hematocrite, thrombocytes) and biochemical tests<br />

(urea, creatinine, bilirubin, natrium, C-reactive<br />

protein, procalcitonin, total blood protein, albumin)<br />

was obtained.<br />

RESULTS: In bleeding patients we found significantly<br />

lower hematocrite (p


(on borderline of statistical significance) in surviving<br />

patients. Higher levels of serum urea and creatinine<br />

were found in patients who died so it probably has an<br />

influence on mortality of bleeding patients.<br />

Abstract 69<br />

Structural Studies of the Hepatitis<br />

C Virus NS5A Protein<br />

R Love, O Brodsky, M Hickey, P Wells, A Zou, W Hao,<br />

R Duggal, and C Cronin<br />

Pfizer Global Research and Development, San Diego, CA,<br />

USA<br />

Background: Non-structural protein NS5A<br />

is a critical component of HCV replication and<br />

is involved in several cellular processes such as<br />

interferon resistance and apoptotic regulation. It is a<br />

phosphoprotein of 447 residues with 3 domains, and<br />

an amphipathic N-terminal helix which promotes<br />

membrane association. Domain I consists of a novel<br />

zinc-binding motif and two subdomains, as shown<br />

by a crystal structure reported in 2005 (Nature,<br />

435:374). That structure revealed a dimer, which in<br />

turn suggested a potential RNA binding cleft. We<br />

have investigated new methods of preparation of<br />

domain 1, with subsequent determination of a novel<br />

crystal structure for the dimer of this protein.<br />

Methods: A novel pET/T7-based HCV NS5A<br />

protein expression vector, utilizing a TEV-cleavable<br />

N-terminal polyhistidine purification tag, was used<br />

to obtain multimilligram quantities of recombinant<br />

NS5A domain I protein (amino acids 33-202). Protein<br />

expression was carried out using Ultra-Yield Flask<br />

technology in Terrific Broth. Soluble NS5A domain I<br />

protein was isolated by passage over Probond IMAC<br />

resin followed by Q-Sepharose chromatography.<br />

The polyhistidine tag was subsequently removed by<br />

cleavage with TEV protease and the protein further<br />

purified by passage over a second ProBond column<br />

followed by size exclusion chromatography. After<br />

crystallization screening to find initial conditions,<br />

Hampton detergent-additives were employed to<br />

optimize crystals of domain 1.<br />

Results: The structure of NS5A domain 1 was<br />

determined to 1.9Å resolution by molecular<br />

replacement, and found to exist as a dimer. The<br />

polypeptide fold within each monomer and the<br />

zinc site are very similar to that reported in 2005,<br />

however, the dimer interface between monomers is<br />

significantly different. In our dimer the long axes of<br />

the monomers are parallel, and related by approximate<br />

2-fold symmetry. The two N-termini are located<br />

on the same end of the dimer, which theoretically<br />

permits membrane association of this end via two<br />

amphipathic N-terminal helices (not present in this<br />

structure). Our dimer interface shows extensive<br />

buried surface area and involves interactions between<br />

a number of conserved residues. Residues defining the<br />

previously proposed RNA-binding cleft now lie fully<br />

exposed and on the side of each monomer farthest<br />

from the dimer interface.<br />

Conclusions: The crystal structure of NS5A<br />

domain 1 that we have determined reveals a mode<br />

of dimerization different from that originally<br />

reported, yet is nevertheless consistent with a<br />

membrane association mechanism. It therefore<br />

offers an alternative possibility for the physiological<br />

configuration of NS5A.<br />

Abstract 70<br />

Dose Selection of Albinterferon<br />

Alfa-2b (alb-IFN) for a Phase 3<br />

Clinical Program<br />

JG McHutchison<br />

Duke Clinical Research Institute and Division of<br />

Gastroenterology, North Carolina, USA<br />

BACKGROUND: alb-IFN, a novel, long-acting<br />

interferon (IFN) for treatment of chronic hepatitis C<br />

(CHC), has demonstrated promising antiviral activity<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 73


and tolerability in preclinical and early clinical studies.<br />

The results of a phase 2b clinical study of alb-IFN were<br />

used to determine dosage regimens for evaluation in<br />

ongoing phase 3 studies.<br />

METHODS: 458 IFN treatment-naïve patients with<br />

genotype 1 CHC were randomized to 4 treatment<br />

arms (peginterferon alfa-2a [PEG-IFNα-2a] 180 μg<br />

qwk or one of 3 alb-IFN arms [900 or 1200 μg q2wk,<br />

or 1200 μg q4wk], all in combination with weightbased<br />

oral ribavirin 1000–1200 mg/d) in an active<br />

controlled, open-label, 48-wk, dose-ranging study.<br />

RESULTS: For alb-IFN 900 μg q2wk (n = 118), the<br />

sustained virologic response (SVR) rate by intent-totreat<br />

analysis was comparable to PEG-IFNα-2a (n =<br />

114): 59% vs 58% (P = NS). The wk-4 and 12 response<br />

rates (defined as hepatitis C virus RNA < limit of<br />

detection, ie,


experiment, the same treatment schedule was use<br />

to determine that the minimal effective CLDC dose<br />

was between 0.5 to 0.05 µg/mouse. CLDC treatment<br />

increased the expression of inflammatory cytokines<br />

IL-1α, MCP-1, RANTES and the T H<br />

1 cytokine IL-12<br />

were statistically increased in the liver the day after<br />

the last treatment. IL-12, MCP-1 and RANTES were<br />

also statistically increased in the serum. To better<br />

understand the temporal expression of the cytokines<br />

in response to CLDC, the serum of C57BL/6 mice at<br />

1, 3, 8 or 24 hr after a single IV injection of CLDC<br />

(5 µg/mouse) was assayed for IFN-α, IFN-γ, IL-6, and<br />

IL-10 (Figure 4). All cytokines were increased with a<br />

peak at 3 hr after CLDC administration, except for IL-<br />

10, which was not affected. The levels of IFN-α, IFN-γ<br />

and IL-6 nearly reached baseline levels by 24 hr.<br />

CONCLUSION: CLDC were effective in reducing liver<br />

HBV DNA probably by non-cytolytic blockage of HBV<br />

DNA replication mediated by IFN-α/β or IFN-γ.<br />

FUNDING: HHSN266200500036C, Enteric and Hepatic<br />

Diseases, NIAID, NIH (JDM)<br />

Abstract 72<br />

Role of Stomatin in the Assembly<br />

of Hepatitis C Virus RNA Replicase<br />

Complex<br />

J-H Kim, DK Ahn, S-B Shim, and J-W Oh<br />

Department of Biotechnology, Yonsei University, 134<br />

Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea<br />

BACKGROUND: Hepatitis C virus (HCV) replication<br />

occurs on a lipid raft or detergent-resistant membrane<br />

(DRM) and is mediated by RNA replicase<br />

complex (RC) consisting of viral nonstructural<br />

proteins including RNA-dependent RNA polymerase<br />

(RdRp), NS5B protein, and cellular proteins. The<br />

DRM structure containing viral RCs can provide a<br />

microenvironment for efficient RNA replication by<br />

concentrating and compartmentalizing the RCs.<br />

Recently, various host proteins interacting with<br />

HCV NS5B have been identified. However, cellular<br />

proteins playing a structural and organizational role<br />

in the viral replication complex formation have not<br />

been yet identified and characterized. Here, we took<br />

a proteomic approach to identify cellular proteins<br />

that might participate in HCV RNA replication<br />

steps including the formation of RCs at cellular viral<br />

replication sites.<br />

METHOD: Cellular proteins interacting with the<br />

NS5B protein were pulled down, subjected to<br />

SDS-PAGE, and identified by mass spectrometry.<br />

Interaction with the NS5B protein was confirmed by<br />

co-immunoprecipitation and immunofluorescence<br />

studies. Cellular localizations of the identified protein<br />

and NS5B protein were assessed by subcellular<br />

fractionation and membrane flotation assays using<br />

the hepatoma stable cell line Huh7 harboring an<br />

HCV subgenomic replicon RNA. Knock-down of<br />

the expression of an NS5B-interacting protein was<br />

carried out using a small hairpin interfering RNA<br />

and an antisense peptide nucleic acid (PNA) specific<br />

to stomatin. Levels of HCV RNA and proteins were<br />

measured by quantitative real-time PCR and Western<br />

blot analysis, respectively.<br />

RESULTS: We identified stomatin as the most<br />

prominent cellular protein interacting with HCV<br />

NS5B. Stomatin has been known to participate<br />

in the formation of lipid rafts, which are<br />

membrane microdomains associated with protein<br />

complexes, cholesterol, and sphingolipids. Coimmunoprecipitation<br />

and co-localization studies<br />

confirmed the in vivo interaction between stomatin<br />

and HCV NS5B, both of which also co-fractionated<br />

with the mitochondria. Membrane flotation assays<br />

showed that these proteins are associated with<br />

the lipid-raft-like domain of the mitochondria. To<br />

characterize the potential role of stomatin in HCV RNA<br />

replication, stoamtin expression was knock-downed<br />

in HCV subgenomic replicon cells using a stomatinspecific<br />

siRNA and PNA. The results indicate that a<br />

decrease of stomatin expression suppresses HCV<br />

RNA replication, which is accompanied with release<br />

of NS5B protein from the DRM to the detergentsensitive<br />

membrane fraction.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 75


CONCLUSIONS: Our results identify stomatin as<br />

a cellular protein involved in HCV RNA complex<br />

formation on a mitochondria-associated detergent<br />

resistant membrane structure.<br />

Abstract 73<br />

Antiviral Activity of Amino Acid<br />

Derivatives of Monascus Pigment<br />

in Hepatitis C Virus Replicating<br />

Cells<br />

S-J Kim, J-H Kim, H Jung, J Jeun, CS Shin, and<br />

J-W Oh<br />

Department of Biotechnology, Yonsei University, 134<br />

Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea<br />

BACKGROUND: Hepatitis C virus (HCV) infection<br />

leads to a serious liver disease and the best present<br />

therapy for chronic hepatitis C is a combination of<br />

pegylated interferon with ribavirin. This therapy<br />

is only effective in half of the patients. Traditional<br />

antiviral drugs designed to target viral enzymes<br />

often generate resistant mutants rapidly. Therefore,<br />

the search for alternative, specific therapies for<br />

HCV chronic infection continues for more options.<br />

Several lines of evidence suggest that cellular lipid<br />

and cholesterol metabolism plays a role directly or<br />

indirectly in the HCV replication cycle. Particularly,<br />

cholesterol biosynthesis has been proposed as an<br />

integral part for HCV replication on lipid rafts and<br />

various inhibitors of 3-hydroxy-3-methylglutaryl<br />

coenzyme A (HMG-CoA) reductase, the key enzyme in<br />

cholesterol biosynthesis, have been shown to suppress<br />

HCV replication. We recently have produced various<br />

amino acid derivatives (AADs) of Monascus pigments<br />

and showed their inhibitory activity against HMG-<br />

CoA reductase. Here, we investigated the potency of<br />

Monascus pigment AADs in HCV replication.<br />

HCV-infected cells were treated with various<br />

concentrations of each AAD alone or in combination<br />

with interferon-alpha (IFN-α). Western blot analyses<br />

and quantitative real-time RT-PCR were carried out to<br />

analyze the levels of HCV nonstructural proteins and<br />

viral RNA in the treated cells. Cytotoxicity of AADs<br />

was evaluated by the MTT assay. Inhibitory activity<br />

of monascus AADs against HMG-CoA reductase was<br />

determined by an enzyme assay measuring NADPH<br />

oxidation level. Cholesterol and lipid levels in mice<br />

fed a high-cholesterol diet without or with AADs were<br />

measured using a commercial enzyme kit. Effect of<br />

AADs on HCV RNA polymerase activity was assessed<br />

by an in vitro polymerase assay with purified HCV<br />

RNA polymerase.<br />

RESULTS: Monascus pigment and some of its AADs<br />

significantly lowered total cholesterol and LDLcholesterol<br />

levels in mice fed a high-cholesterol<br />

diet. The pigments also showed various degrees of<br />

inhibitory effect on HMG-CoA reducatse. Treatment<br />

of HCV subgenomic replicon cells with AADs reduced<br />

the expression levels of HCV nonstructural proteins<br />

and resulted in suppression of HCV RNA replication.<br />

Furthermore, combination of AADs with IFN-α<br />

potentiated the anti-HCV activity of IFN-α with no<br />

significant increase in cytotoxicity. Similar antiviral<br />

activity was also observed in HCV genotype 2a<br />

JFH1-infected Huh7 cells. Finally, we observed no<br />

strict correlation between the inhibitory activity of<br />

AADs against HMG-CoA reducatse and their anti-<br />

HCV activity, raising a possibility that alternative<br />

mechanisms could explain the antiviral activity of<br />

AADs.<br />

CONCLUSIONS: Our results identify Monascus<br />

pigment AADs as a potential inhibitor of HCV<br />

replication and suggest that combination with INF-α<br />

or possibly with other anti-HCV drugs might offer<br />

an alternative strategy with which to control HCV<br />

replication.<br />

METHODS: The hepatoma stable cell line supporting<br />

autonomous replication of a genotype 1b HCV<br />

subgenomic replicon RNA and the genotype 2a<br />

76 Global Antiviral Journal Volume 3, Supplement 2


Abstract 74<br />

Silymarin Displays Anti-Viral<br />

Anti-Inflammatory, and<br />

Immunomodulatory Effects<br />

Towards Hepatitis C Virus<br />

J Wagoner 1 , C Morishima 1 , O Kane 1 , D Lee 2 , and<br />

S Polyak 1<br />

1 University of Washington, Seattle, WA, USA; 2 Harvard<br />

University, Cambridge, MA, USA<br />

BACKGROUND: A striking feature of hepatitis C<br />

virus (HCV) infection is its propensity to establish a<br />

chronic disease state. Because state-of-the art antiviral<br />

treatments are costly, have serious side-effect<br />

profiles, and moderate probabilities for durable<br />

cures, many patients opt for complementary and<br />

alternative medicine (CAM)-based approaches to<br />

improve liver health. However, the mechanisms of<br />

hepatoprotection have not been characterized.<br />

METHODS: In the current study, we examined<br />

silymarin, derived from milk thistle, for anti-viral,<br />

anti-inflammatory and immunomodulatory effects<br />

towards hepatitis C virus.<br />

RESULTS: Silymarin inhibited expression of TNF-α<br />

in anti-CD3 stimulated human PBMC and NF-κB<br />

dependent transcription in T cells and in human<br />

hepatoma Huh7 cells. Silymarin also caused dosedependent<br />

inhibition of infection of Huh7 and<br />

Huh7.5.1 cells by JFH-1 virus, with both prophylactic<br />

and therapeutic effects. When combined with IFN-α,<br />

Silymarin inhibited HCV replication more than<br />

IFN-α alone. Anti-viral effects induced by Silymarin<br />

involved Jak-Stat dependent and independent<br />

signaling. These activities were observed with an<br />

independently standardized preparation as well as 4<br />

commercial sources of silymarin. HPLC fractionation<br />

of the botanical medicine permitted identification<br />

of the components eliciting anti-viral and antiinflammatory<br />

actions.<br />

CONCLUSIONS: The data demonstrate that standardized<br />

silymarin has anti-viral action against in<br />

vitro HCV infection. Furthermore, this botanical<br />

medicine also displays potent immunomodulatory<br />

and anti-inflammatory actions. Therefore, CAMbased<br />

approaches may assist in the management<br />

of patients with chronic hepatitis C. Identification<br />

of the bioactive molecules in Silymarin provides<br />

an opportunity for rationale drug design through<br />

medicinal chemistry approaches.<br />

Abstract 75<br />

Construction and Applications of<br />

a Liver-specific Lentivirus Vector<br />

with Host Range Determined by<br />

the Envelope Proteins of HBV<br />

N Chai, H Chang, E Nicolas, S Gudima, and J Taylor<br />

Fox Chase Cancer Center, Pennsylvania, USA<br />

BACKGROUND: There is a pressing need for liver<br />

specific vectors for treatment of genetic diseases<br />

affecting the liver and for therapy of chronic hepatic<br />

infections (e.g., by HCV). Construction of lentivirus<br />

vectors with a liver specific host range would be an<br />

important step towards these goals. HBV is highly<br />

liver specific, targeting hepatocytes and possibly<br />

bile duct epithelial cells. Therefore, if the envelope<br />

proteins of HBV could be presented on the surface of<br />

a lentivirus vector, liver-specific gene delivery should<br />

be achieved. We describe a vector that appears to<br />

fulfill this need, with an in vitro host range that is<br />

restricted to primary human hepatocyte cultures.<br />

METHODS: A combination of plasmids was used<br />

to transfect 293 cells, with the aim of making an<br />

HIV-1 pseudotype with the envelope proteins L and<br />

S of HBV. The pseudotyped HIV, HIV(LS), was tested<br />

for the ability to infect cultures of primary human<br />

hepatocytes (PHH) as well as a number of different<br />

cell lines. Infection was detected by the expression of<br />

a reporter gene, LacZ, either using X-gal staining or<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 77


eal-time PCR for detection of LacZ mRNA. Vector<br />

uptake was compared to uptake of HBV and HDV<br />

using inhibitors previously reported to distinguish<br />

forms of receptor mediated endocytosis.<br />

RESULTS: HIV(LS) demonstrated the same in vitro<br />

host range as HBV and HDV, infecting PHH but not<br />

primary woodchuck hepatocyes, or established cell<br />

lines such as Huh7, HepG2, and 293 cells. Staining<br />

for LacZ expression revealed that ~10% of the cells<br />

in a typical preparation of PHH could be infected,<br />

similar to the infection efficiency of HDV as assessed<br />

by staining for delta antigen. However, the uptake<br />

of HIV(LS) appeared to involve a different pathway<br />

than either HBV or HDV. HBV and HDV behaved as<br />

if entry required an endosomal mechanism coupled<br />

with acidification. HIV(LS) appeared to enter cells<br />

by direct fusion of the virus particles at the plasma<br />

membrane.<br />

CONCLUSIONS: We have constructed a lentivirus<br />

vector that will specifically and efficiently infect<br />

human hepatocytes. It will find application in liverspecific<br />

gene therapies, including what might be an<br />

efficient one-shot vaccine against HBV. In addition,<br />

although the lentivirus uses the same envelope<br />

proteins as HBV and HDV, for attachment and entry,<br />

it uses them in a significantly different way.<br />

Abstract 76<br />

Designed Zinc Finger Proteins<br />

Bind Duck Hepatitis B Virus<br />

Enhancer DNA and Decrease<br />

Production of Viral RNA, Proteins<br />

and Progeny In Vitro<br />

KA Zimmerman, KP Fischer, MA Joyce, and<br />

DLJ Tyrrell<br />

University of Alberta, Edmonton AB, Canada<br />

BACKGROUND: Duck hepatitis B virus (DHBV) is<br />

a model virus for human hepatitis B virus (HBV),<br />

which persistently infects approximately 360 million<br />

individuals worldwide. Nucleoside analogs such as<br />

lamivudine, tenofovir and adefovir inhibit the viral<br />

polymerase and decrease virus production, however,<br />

due to persistence of the HBV episome, none<br />

completely clear the virus in most treated patients.<br />

HBV DNA is present in the nucleus as a closed<br />

covalently circular (cccDNA) form, where it drives<br />

viral transcription and virus production. cccDNA is<br />

the next target for therapeutics aiming to clear the<br />

viral infection entirely.<br />

METHODS: To specifically target cccDNA, we have<br />

designed zinc finger proteins (ZFPs) that bind<br />

to DNA in the DHBV enhancer region. We have<br />

designed and purified two ZFPs targeting 18bp<br />

sequences and two pairs of ZFPs each targeting 9bp<br />

sequences. Binding kinetics were assessed using<br />

surface plasmon resonance (SPR) and electrophoretic<br />

mobility shift assays (EMSA). In vitro pull-down<br />

assays were used to demonstrate the ability of ZFPs<br />

to bind authentic cccDNA. The ZFPs were cloned into<br />

a mammalian expression vector and co-transfected<br />

into LMH (chicken hepatoma) cells with the plasmid<br />

pDHBV1.3, which replicates the DHBV replication<br />

cycle. Transfected cells were analyzed by Western blot<br />

on whole cell lysates, Southern blot on intracellular<br />

viral (ICV) DNA and quantitative PCR on Trizol<br />

isolated RNA to investigate the effects of ZFPs on<br />

viral products. Cell viability after transfection was<br />

assessed using an MTT assay.<br />

78 Global Antiviral Journal Volume 3, Supplement 2


RESULTS: Using SPR and EMSA, we determined the<br />

dissociation constants (Kd) to be in the nanomolar<br />

range (12.3-99nM) for four ZFPs, in the picomolar<br />

range (471pM) for one ZFP and the micromolar<br />

range (185uM) for the last ZFP. The in vitro pulldown<br />

assay demonstrated that the ZFPs could bind<br />

authentic cccDNA. Western blots for DHBV proteins<br />

on LMH cell lysates showed dramatic decreases in<br />

viral core and surface expression in the presence of<br />

all ZFPs compared to actin controls. Quantitative<br />

PCR for viral core, polymerase and surface RNA<br />

levels demonstrated significant decreases of 58-88%,<br />

compared to co-transfection with empty vector. In<br />

addition, ICV particle production was decreased in<br />

ZFP-transfected cells.<br />

CONCLUSION: Our designed ZFPs can bind target<br />

sequences with Kd’s in ranges adequate for drug<br />

development. They are able to bind authentic cccDNA<br />

in vitro and can decrease the production of viral<br />

products in the LMH tissue culture system. We suggest<br />

our designed ZFPs are binding the DHBV enhancer in<br />

vitro and preventing the transcriptional machinery<br />

from accessing it, resulting in decreased production<br />

of viral mRNA, proteins and genomic RNA.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 79


Resistance to<br />

Antiviral Agents<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 81


Abstract 77<br />

Overcoming HCV Treatmentresistant<br />

Characteristics<br />

MW Fried<br />

University of North Carolina at Chapel Hill, USA<br />

The response to peginterferon and ribavirin, while<br />

effective for the majority of patients, is neither<br />

universal nor guaranteed and is highly related to<br />

multiple virologic and host factors 1-3 . Genotype 1<br />

and baseline levels of HCV RNA (above 400,000 IU/<br />

mL) are the strongest virological factors associated<br />

with diminished response rates 1-3 . Among host<br />

characteristics, the presence of cirrhosis, African<br />

American race, obesity, insulin resistance and<br />

hepatic steatosis have also been demonstrated to<br />

decrease the rate of SVR 1, 2, 4-6 . Although it is expected<br />

that newer classes of drugs, such as protease and<br />

polymerase inhibitors, will improve sustained<br />

response rates for all individuals, these agents remain<br />

investigational. Furthermore, it has become apparent<br />

that peginterferon and ribavirin will serve as the<br />

backbone of triple drug regimens for the foreseeable<br />

future 7, 8 . Therefore, optimizing peginterferon and<br />

ribavirin remains an important goal.<br />

The mechanism by which increased body weight<br />

diminishes antiviral response, regardless of<br />

peginterferon preparation, is likely complex and<br />

multifactorial. Obesity, a surrogate marker for<br />

hepatic steatosis, has been associated with decreased<br />

antiviral efficacy as well as increased risk of fibrosis,<br />

while hepatic steatosis is also frequently associated<br />

with insulin resistance that impairs antiviral and<br />

immune-stimulating properties of peginterferon-alfa<br />

(5,10–12).<br />

Individual patients with chronic hepatitis C represent<br />

a blend of multiple attributes that can affect antiviral<br />

response. Thus, a patient with genotype 1 infection<br />

may also have high levels of viremia and be overweight,<br />

a combination of factors whose pretreatment<br />

probability of response has been shown to be lower<br />

than any single unfavorable predictive factor 9 . These<br />

poor prognostic factors tend to cluster together,<br />

defining a population of patients who are least likely<br />

to respond to conventional antiviral therapy with<br />

peginterferon and ribavirin 10 .<br />

Numerous strategies have been suggested to improve<br />

the SVR rates for patients predicted to be poorly<br />

responsive to interferon-based therapies. Increasing<br />

the dose of peginterferon, increasing the dose of<br />

ribavirin and prolonging the duration of therapy have<br />

all met with limited success 11-14 . The use of higher<br />

weight-based ribavirin doses in combination with<br />

peginterferon alfa-2b produced a significant, albeit<br />

modest, improvement in a large community-based<br />

study 15 . A recent study compared the impact on viral<br />

kinetics and SVR of intensified treatment regimens<br />

of peginterferon alfa-2a and ribavirin with standard<br />

regimens in patients with a cluster of poor prognostic<br />

factors (genotype 1, high baseline levels of HCV RNA<br />

and body weight >85 kg) 16 . The results demonstrated<br />

that it is possible to improve virologic response rates<br />

by increasing the intensity of treatment 16 .<br />

Peginterferon and ribavirin are important components<br />

of antiviral therapy for chronic hepatitis C. In<br />

combination with direct antivirals, fewer patients<br />

will demonstrate treatment-resistant characteristics<br />

although optimizing response to peginterferon and<br />

ribavirin will remain essential.<br />

References:<br />

1. Fried MW, Shiffman ML, Reddy KR, et al.<br />

Combination of peginterferon alfa-2a plus<br />

ribavirin in patients with chronic hepatitis C<br />

virus infection. New England Journal of Medicine<br />

2002;347:975-82.<br />

2. Manns MP, McHutchison JG, Gordon SC, et al.<br />

Peginterferon alfa-2b plus ribavirin compared<br />

with interferon alfa-2b plus ribavirin for initial<br />

treatment of chronic hepatitis C: a randomised<br />

trial. Lancet 2001;358:958-65.<br />

3. Hadziyannis SJ, Sette H, Jr., Morgan TR, et al.<br />

Peginterferon-alpha2a and ribavirin combination<br />

therapy in chronic hepatitis C: a randomized<br />

study of treatment duration and ribavirin dose.<br />

Ann Intern Med 2004;140:346-55.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 83


4. Conjeevaram HS, Fried MW, Jeffers LJ, et al.<br />

Peginterferon and ribavirin treatment in African<br />

American and Caucasian American patients<br />

with hepatitis C genotype 1. Gastroenterology<br />

2006;131:470-7.<br />

5. Zeuzem S, Hultcrantz R, Bourliere M, et al.<br />

Peginterferon alfa-2b plus ribavirin for treatment<br />

of chronic hepatitis C in previously untreated<br />

patients infected with HCV genotypes 2 or 3. J<br />

Hepatol 2004;40:993-9.<br />

6. Dev A, Patel K, McHutchison JG. Hepatitis C and<br />

steatosis. Clin Liver Dis 2004;8:881-92.<br />

7. Kieffer T, Sarrazin C, Miller J, al e. Combination<br />

of telaprevir and peg-IFN alfa suppresses both<br />

wild-type virus and resistant variants in HCV<br />

genotype-1 infected patients in a 14-day phase<br />

1b study. Hepatology 2006:Abstract 92.<br />

8. Tong X, Chase R, Skelton A, Chen T, Wright-<br />

Minogue J, Malcolm BA. Identification and<br />

analysis of fitness of resistance mutations against<br />

the HCV protease inhibitor SCH 503034. Antiviral<br />

Res 2006;70:28-38.<br />

9. Foster GR, Fried MW, Hadziyannis SJ, Messinger<br />

D, Freivogel K, Weiland O. Prediction of sustained<br />

virological response in chronic hepatitis C patients<br />

treated with peginterferon alfa-2a and ribavirin.<br />

Scandinavian Journal of Gastroenterology<br />

2007;42:247-55.<br />

10. Swain M, Foster GR, Hadziyannis SJ. Poor<br />

prognostic factors in heavier patients with chronic<br />

hepatitis C result in less favourable treatment<br />

outcomes. Journal of Hepatology 2006;44:227<br />

(abstract).<br />

11. Sanchez-Tapias JM, Diago M, Escartin P, et<br />

al. Peginterferon-alfa2a plus ribavirin for 48<br />

versus 72 weeks in patients with detectable<br />

hepatitis C virus RNA at week 4 of treatment.<br />

Gastroenterology 2006;131:451-60.<br />

12. Lodato F, Azzaroli F, Brillanti S, et al. Higher doses<br />

of peginterferon alpha-2b administered twice<br />

weekly improve sustained virological response in<br />

difficult-to-treat patients with chronic hepatitis<br />

C: results of a pilot randomized study. J Viral<br />

Hepat 2005;12:536-42.<br />

13. Bacon BR, et al. The DIRECT Trial (Daily-Dose<br />

Consensus Interferon and Ribavirin: Efficacy<br />

of Combined Therapy): Treatment of Non-<br />

Responders to Previous Pegylated Interferon plus<br />

Ribavirin: Sustained Virologic Response Data.<br />

Hepatology 2007:(abstract).<br />

14. Marcellin P, et al. Pegylated interferon alfa-<br />

2a (40KD) plus ribavirin (RBV) in prior nonresponders<br />

to pegylated interferon alfa-2b<br />

(12KD)/RBV: final efficacy and safety outcomes of<br />

the REPEAT study Hepatology 2007:(abstract).<br />

15. Jacobson IM, Brown RS, Freilich B, al<br />

e. Peginterferon alfa-2b and weight-based or flatdose<br />

ribavirin in chronic hepatitis C: a randomized<br />

trial. Hepatology 2007;46:971-81.<br />

16. Fried MW, Jensen D, Rodriguez-Torres M, et al.<br />

Improved outcomes in HCV patients with difficult<br />

to treat characteristics: randomized study of<br />

Intensified peginterferon α-2a and ribavirin.<br />

Hepatology (abstract) 2006.<br />

Abstract 78<br />

Genetic and Structural Variability<br />

of the Hepatitis C Viral<br />

Polymerase, NS5B: Implications<br />

for Resistance to Inhibitors<br />

PC Simister 1 , R Brillet 2 , V Lohmann 3 , J-M Pawlotsky 2 ,<br />

and S Bressanelli 1<br />

1<br />

Laboratoire de Virologie Moléculaire et Structurale,<br />

CNRS, Gif-sur-Yvette, France; 2 Hôpital Henri Mondor,<br />

Créteil, France; 3 Department of Molecular Virology,<br />

University of Heidelberg, Heidelberg, Germany<br />

The HCV polymerase, NS5B, is an important target<br />

in the development of anti-HCV drugs. Since 1999,<br />

40 NS5B crystal structures have been solved and<br />

deposited in the Protein Data Bank (PDB). Of these,<br />

36 structures are based on genotype-1b sequences<br />

and 4 on genotype 2a. Furthermore, in our laboratory<br />

we have crystallised NS5B from the JFH1 strain<br />

(genotype 2a). The catalytic region of NS5B consists<br />

of three domains (thumb, palm and fingers), which<br />

are connected to a transmembrane helix at the<br />

C-terminus by a flexible linker. Significant domain<br />

84 Global Antiviral Journal Volume 3, Supplement 2


movements are expected to occur during the different<br />

stages of polymerisation of the HCV RNA genome.<br />

However, all the structures published to date have<br />

been crystallised in very similar conformations.<br />

The available structures represent predominantly<br />

two truncated forms in which either the membrane<br />

anchor is deleted (delta-21) or both the anchor and<br />

the linker (delta-55).<br />

Part 1: After analysing the slight conformational<br />

differences between the available structures<br />

using computational methods, we found that the<br />

conformationally invariant regions are not necessarily<br />

identical between genotypes. Also, in the structures<br />

from genotype 1b, the more open conformation is<br />

seen only in the truncated delta-55 form. However,<br />

for genotype 2a both conformations are observed with<br />

the delta-21 truncation. Significantly, we discovered<br />

that global conformational changes appear not to<br />

occur upon inhibitor binding. Interestingly, a detailed<br />

comparison of models of the JFH1 and other 2a<br />

polymerases has provided structural insights helping<br />

to explain their different biological activities.<br />

Part 2: Consensus genotype-1b sequences were<br />

obtained from the HCV-genome database, euHCVdb<br />

(Combet et al., 2007). Additionally, we characterized<br />

the quasi-species in 2 untreated patients infected<br />

with HCV (genotype 1b) and obtained about 40 fulllength<br />

NS5b sequences. We analysed the location<br />

of polymorphisms in the consensus sequences and<br />

those from the clinical samples in order to understand<br />

how inherent mutability might affect the clinical<br />

application of anti-NS5B inhibitors. Many residues in<br />

the inhibitor-binding sites are polymorphic including<br />

those which correspond to known resistance<br />

mutations. Importantly, some polymorphisms in the<br />

quasi-species are not represented in the consensus<br />

sequences, implying that extra information is<br />

provided by the clinical samples.<br />

Thus, we confirm that the C-terminal region is<br />

essential for NS5B regulation. The structural<br />

differences observed might affect the efficacy of<br />

anti-NS5B inhibitors in a genotype-dependent<br />

manner. Furthermore, the presence of pre-existing<br />

polymorphisms associated with resistance in the<br />

quasi-species population may influence the speed of<br />

HCV rebound with important implications for the<br />

design of therapeutic strategies.<br />

Abstract 79<br />

Selection of Multidrug Resistant<br />

Hepatitis B Virus Following<br />

Sequential Monotherapy and<br />

Combination Therapy<br />

A Ayres 1 , A Bartholomeusz 1 , M Littlejohn 1 , D Colledge 1 ,<br />

L Yuen 1 , P Angus 2 , A Thompson 1 , and S Locarnini 1<br />

1 Victorian Infectious Diseases Reference Laboratory, Nth<br />

Melbourne, Victoria, Australia; 2 Austin and Repatriation<br />

Medical Centre, Heidelberg, Victoria, Australia<br />

Background: Although more antiviral agents for<br />

treatment of hepatitis B virus (HBV) infection have<br />

become available in recent years, options remain<br />

limited. Currently they comprise conventional and<br />

pegylated interferons and four oral nucleos(t)ides<br />

analogues. The former show limited efficacy whilst<br />

long-term use of the latter may cause the emergence<br />

of drug resistant HBV; their sequential use may result<br />

in multidrug resistant HBV variants, which are posing<br />

an increasing clinical challenge.<br />

Aim: To perform clonal analyses of serial putative<br />

drug resistant HBV isolates in order to monitor the<br />

emergence of multidrug resistance in relationship to<br />

quasispecies diversity.<br />

Methods: The HBV polymerase gene was amplified,<br />

cloned and sequenced from HBV isolates obtained<br />

from four different patients. Patient A initially failed<br />

adefovir monotherapy and subsequently failed to<br />

respond to combination therapy with adefovir (ADV)<br />

and lamivudine (LMV). Patients B and C were already<br />

infected with LMV resistant mutants and also failed<br />

to respond to ADV and LMV in combination. HBV<br />

infection in patient D became sequentially refractory<br />

to monotherapy with LMV, entecavir (ETV) and<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 85


ADV and subsequently became unresponsive to a<br />

combination of ADV and ETV.<br />

Results and Discussion: Direct sequencing<br />

and clonal analysis of HBV isolates from samples<br />

from all four patients revealed complex mutation<br />

profiles. Sequencing of HBV from Patient A revealed<br />

the presence of rtM250L + rtI233V substitutions<br />

in addition to those (rN236T + rtA181T) known to<br />

confer ADV resistance. Clonal analysis of HBV from<br />

patients B and C revealed the co-existence of two<br />

separate HBV populations, each of which encoded<br />

different primary resistance mutations. Lamivudine<br />

resistant mutants (encoding rtL180M+ rtM204V/I)<br />

comprised one population, whilst a second population<br />

harboured mutations encoding rN236T +/- rtA181V,<br />

which confer ADV resistance. Clones that encoded<br />

both rtN236T and rtM204I/V on the same genome<br />

were not detected. Isolates from patient D encoded<br />

a previously unreported variant (rtN236V) in<br />

association with changes that confer primary LMV<br />

resistance (rtL180M + rtM204V) and primary<br />

ETV resistance (rtL180M + rtT184G + rtM204V+<br />

rtS202I).<br />

Conclusion: Multidrug resistant HBV mutants<br />

that encode complex changes in the viral polymerase<br />

are emerging in patients who have undergone<br />

sequential antiviral therapy. Resistance to more<br />

than one nucleoside inhibitor is a major concern and<br />

demonstrates the need for continuous surveillance.<br />

Monitoring the evolution of HBV mutants selected by<br />

during different treatment regimens will contribute<br />

to the development of optimal treatment regimes<br />

tailored to individual patients as well as contributing<br />

to our understanding of the factors that contribute to<br />

drug resistance.<br />

Abstract 80<br />

Multidrug Resistance and Crossresistance<br />

Pathways in HBV as a<br />

Consequence of Treatment Failure<br />

L Yuen 1,2 , A Ayres 1 , A Bartholomeusz 1 , M Littlejohn 1 ,<br />

and S Locarnini 1<br />

1 VIDRL, North Melbourne, Australia; 2 Evivar Medical,<br />

East Melbourne, Australia<br />

Background/Aims: In recent years, the sequential<br />

use of anti-viral medications for the treatment of<br />

hepatitis B has lead to the emergence of complex<br />

multi drug resistant HBV. Primary drug resistance<br />

mutations result in reduced susceptibility to an<br />

antiviral agent, while secondary compensatory<br />

mutations restore replication defects associated with<br />

primary drug resistance, and may be associated with<br />

low-level reduced susceptibility. Several evolutionary<br />

pathways of drug resistance for HBV have been<br />

observed in patients treated with (nucleot(s)ides<br />

NAs. It is possible that the drug resistance mutations<br />

selected under one agent may affect the efficacy<br />

of subsequent agents. The aim of this study was<br />

to elucidate mutation pathways associated with<br />

resistance to NAs and to determine the potential<br />

cross-resistance profiles selected under a particular<br />

NA.<br />

Methods: The HBV reverse transcriptase (rt) gene<br />

was sequenced from patients pre-therapy and during<br />

virological breakthrough on antiviral therapy. A HBV<br />

sequence analysis program, SeqHepB, was used to<br />

analyse the treatment-associated mutations. The<br />

sequence data obtained from the most recent samples<br />

of 159 pre therapy patients and 215 patients during<br />

their virological breakthrough were compared while<br />

receiving lamivudine (LMV) and/or adefovir (ADV).<br />

Associations were evaluated using the Fisher’s Exact<br />

method and a pattern discovery program Magnum<br />

Opus.<br />

Results and Discussion: Treatment with LMV,<br />

L-nucleoside analogues (such as telbivudine (LdT)),<br />

86 Global Antiviral Journal Volume 3, Supplement 2


and/or entecavir (ETV) can result in the selection of<br />

the mutation rtM204I/V. Statistical analysis identified<br />

the selection of some secondary compensatory<br />

mutations (rtL80I/V, rtV173L and rtL180M) during<br />

LMV monotherapy that can also affect response<br />

to ETV or LdT. In contrast, ADV treatment failure<br />

is typically associated with the selection of the<br />

mutations rtN236T and/or rtA181V/T. During ADV<br />

monotherapy the presence of the mutations rtA181T/V<br />

and/or rtN236T were statistically significant. The<br />

selection of rtA181V and/or rtN236T and loss of<br />

rtL180M and/or rtM204I/V was also statistically<br />

significant among LMV resistant patients who were<br />

subsequently switched to ADV monotherapy. The<br />

rtA181T mutation has been implicated with reduced<br />

sensitivity to LMV and ADV, thus it is common to<br />

both the “204 and 236 pathways”.<br />

Conclusion: Current emerging patterns of antiviral<br />

drug resistance in the HBV polymerase are<br />

complex and codon analysis has identified a number<br />

of amino acids which have occurred at statistically<br />

significant levels while under drug selection<br />

pressure.<br />

Three major HBV evolutionary NA-resistance<br />

pathways (rtM204I/V, rtN236T and rtA181T/V)<br />

have been characterised. The first two pathways are<br />

associated with clusters of secondary mutations that<br />

can affect subsequent treatment with NAs, whilst the<br />

third rtA181T/V is inherently a multi-drug resistance<br />

pathway.<br />

Abstract 81<br />

Antiviral Effect of Nucleoside<br />

Analogs and Interferon on a<br />

Novel Infectious Cloned Hepatitis<br />

C Virus Containing the S282T<br />

Mutation in the NS5B RNA<br />

Polymerase<br />

G Mateu 2 , L Bassit 1,3 , A Grakoui 2 , and RF Schinazi 1,2,3<br />

1 Emory University/VA Medical Center, Department of<br />

Pediatrics, Decatur, GA, USA; 2 Emory University/Yerkes,<br />

Microbiology and Immunology, Atlanta, GA, USA;<br />

3 Veterans Affairs Medical Center, Decatur, GA, USA<br />

BACKGROUND: A chimeric HCV genotype 2a<br />

infectious clone (C-p7) that infects hepatoma cells<br />

and produces a high level of infectious particles in<br />

Huh7.5 cell line was recently developed (Mateu et<br />

al., HCV 2007, Glasgow). In this in vitro C-p7 acute<br />

infectious system, HCV replication was inhibited by<br />

several 2’-Me nucleosides, and interferon alpha-2b<br />

(IFN) in a dose-response manner (Schinazi et al., HCV<br />

2007, Glasgow).<br />

METHODS: To assess resistance to antiviral agents,<br />

cloned virus containing the S282T mutation<br />

associated with 2’-Me nucleoside analog was created.<br />

Activity and toxicity of seven anti-HCV nucleoside<br />

analogs, IFN, and ribavirin (RBV) were evaluated<br />

with this new infectious cloned virus and compared<br />

to wild type HCV. All compounds except ribavirin<br />

are selective anti-HCV agents in the HCV (clone B)<br />

replicon. Following five days incubation, total cellular<br />

RNA was extracted and C-p7 viruses (wild type and<br />

S282T mutant), and ribosomal RNA were amplified.<br />

RESULTS: The C-p7 S282T point mutation in the<br />

coding sequence of NS5B conferred marked resistance<br />

to six of the seven 2’-C-methyl nucleosides evaluated.<br />

This mutation conferred a high level of resistance<br />

(10-25-fold increase in EC 90<br />

) to four compounds<br />

including 2’-C-MeC, NM107 and other 2’-methylated<br />

nucleosides. In contrast, 2’-F-C-MeC, PSI-6130<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 87


showed only a 1.4-fold increase at the EC 90<br />

level. All<br />

compounds, except 2’-C-MeA (CC 50<br />

= 10 µM), showed<br />

no cytotoxicity against the C-p7 S282T mutant. As<br />

expected, IFN showed activity against the C-p7 S282T<br />

mutant, and RBV at a non-toxic concentration (< 30<br />

µM) showed minimal activity (EC 90<br />

= 23 µM).<br />

CONCLUSIONS: Cell-based assays with the full-length<br />

infectious C-p7 viruses have provided powerful and<br />

robust tools for developing new anti-HCV agents that<br />

are not cross-resistant. These results suggest that<br />

the genotype 2a S282T variant of full-length NS5B<br />

represents effective model for resistance profiling of<br />

NS5B polymerase inhibitors.<br />

Abstract 82<br />

Understanding the Molecular<br />

Basis of HBV Drug Resistance by<br />

Molecular Modeling<br />

A Sharon and CK Chu<br />

The University of Georgia College of Pharmacy, Athens,<br />

GA 30602, USA<br />

BACKGROUND: Despite the significant successes in<br />

the treatment of chronic hepatitis B, infection, the<br />

development of resistance against available therapy<br />

is a critical issue. A recent studies show the clinical<br />

frequency of 3TC-resistant mutants is 7.9% for<br />

L180M, 28.6% for M204I, 22.2% for L180M/M204I<br />

and 41.3% for L180M/M204V. The aim of present<br />

investigation is to understand the molecular basis<br />

of drug resistance conferred by the B and C domain<br />

mutations on the binding mode of five clinically<br />

effective anti-HBV agents [lamivudine (3TC),<br />

adefovir (ADV), entecavir (ETV), telbivudine (LdT),<br />

and clevudine (L-FMAU)].<br />

METHODS: Absence of the crystal structure of<br />

HBV RT prompted us to use the homology modeled<br />

structure of HBV RT to gain insight of the drug<br />

resistance mechanism. Minimization, conformational<br />

search and induced fit docking followed by binding<br />

energy calculation on wild type as well as on<br />

mutant-HBV polymerase (L180M, M204V, M204I,<br />

L180M+M204V, L180M+M204I) were performed.<br />

RESULTS: A significant correlation has been observed<br />

between the fold resistance (FR: based on IC 50<br />

values)<br />

and the binding affinity of the nucleoside analogs.<br />

Binding mode studies reveals that the domain C<br />

residue M204 is closely associated with sugar/<br />

pseudosugar ring positioning in the active site. The<br />

positioning of oxathiolane ring of 3TC is plausible<br />

due the induced fit orientation of the M204 residue<br />

in wild type, and further mutation of M204 to V204<br />

or I204 reduces the final binding affinity, which leads<br />

to the drug resistance. The domain B residue L180 is<br />

not directly interact with the nucleoside analogs, but<br />

indirectly associated with other hydrophobic residues<br />

such as Ala87, Phe88, P177 and M204. These five<br />

hydrophobic residues can directly affect the incoming<br />

nucleoside analog in terms of its association and<br />

interaction that can alter the final binding affinity<br />

of the molecule. There was no sugar ring shifting<br />

observed in the case of adefovir and entecavir, while<br />

exocyclic double bond of entecavir oriented to the<br />

backside small hydrophobic pocket (made by residues<br />

A87, F88, P177, L180 and M204) and enhances the<br />

binding affinity. LdT and L-FMAU showed backward<br />

shifting along with upward movement of sugar ring<br />

without enforcing M204 residue for induced fit<br />

movement which makes these molecules competitive<br />

inhibitor of HBV-RT, without being incorporated into<br />

the growing HBV-DNA chain.<br />

CONCLUSION: Modeling results reveal the additive<br />

nature of the dual mutation (L180M + M204V/I)<br />

causing L-nucleosides reduced binding in comparison<br />

to the single mutation. Structural information<br />

obtained from our molecular modeling studies of<br />

nucleosides aids to gain insight of the molecular basis<br />

of drug resistance of anti-HBV agents, which may<br />

be utilized for the future discovery of new anti-HBV<br />

agents (Supported by NIH AI25899).<br />

88 Global Antiviral Journal Volume 3, Supplement 2


Abstract 83<br />

High Genetic Barrier to HCV<br />

Resistance Presented by PSI-6130<br />

A Uzgiris 1 , H Micolochick Steuer 2 , E Murakami 2 , H Bao 2 ,<br />

S Cruz 1 , AG Shields 1 , S Ali 3 , WR Jiang 3 , J Symons 3 ,<br />

PA Furman 2 , and A De La Rosa 2<br />

1 Siemens Medical Solutions Diagnostics, Berkeley, CA,<br />

USA; 2 Pharmasset Inc, Princeton, NJ, USA; 3Roche Palo<br />

Alto LLC, Palo Alto, CA, USA<br />

BACKGROUND: PSI-6130 (β-D-2′-deoxy-2′-fluoro-<br />

2′-C-methylcytidine) is the parent molecule of the<br />

prodrug R7128, an orally active inhibitor of the<br />

hepatitis C virus NS5B polymerase currently in clinical<br />

development. To date, there has been no report of<br />

the emergence of viral resistance to R7128 in clinical<br />

studies. However, reduced activity was seen with<br />

PSI-6130 when tested against a replicon containing<br />

the S282T mutation in NS5B. We have analyzed a<br />

proprietary database of more than 2,800 HCV NS5B<br />

sequences representing six HCV genotypes for the<br />

presence of the S282T substitution.<br />

METHODS: More than 2,800 HCV NS5b sequences<br />

from public databases and from databases at Siemens<br />

were combined for analysis. HCV-positive clinical<br />

sample data were generated with the Siemens<br />

TRUGENE® HCV 5′NC Genotyping Kit* and the HCV<br />

NS5b Assay offered through the Siemens Medical<br />

Solutions Diagnostics Clinical Laboratory. Sequences<br />

from the Los Alamos HCV database were analyzed for<br />

redundancy and degenerate bases. Unique sequences<br />

without ambiguities were selected and subtypes<br />

were confirmed by phylogenetic analysis. Sequential<br />

passaging experiments were performed with Huh7<br />

cells harboring the GT-1b wild-type replicon. Enzyme<br />

inhibition studies were performed with wild-type and<br />

mutant HCV RdRp and the two active triphosphate<br />

metabolites of PSI-6130: PSI-6130-TP and the<br />

triphosphate of the uridine metabolite RO2433.<br />

substitution at position 282 was observed in less than<br />

1% of the sequences analyzed. The two substitutions<br />

observed were the S282T (ACC and ACT) and S282R<br />

(AGG and CGA). The S282T substitution results<br />

in only a 3.8-fold reduced sensitivity to PSI-6130.<br />

In vitro studies showed that HCV replicons containing<br />

the S282T mutation were selected by long-term<br />

passaging (6–8 months) in the presence of PSI-6130.<br />

The S282T substitution reduced the replication<br />

capacity to 15-20% of the wild-type replicon. PSI-<br />

6130-TP was a more potent inhibitor of the wild-type<br />

and S282T mutant polymerases than was RO2433-<br />

TP.<br />

CONCLUSIONS: The S282T mutation that confers<br />

reduced sensitivity to PSI-6130 results in a small<br />

loss in antiviral activity and the substitution is not<br />

highly polymorphic in the wild-type HCV population.<br />

These in vitro data indicate that there is a high genetic<br />

barrier to resistance to PSI-6130. Since PSI-6130 is<br />

metabolized to two active triphosphate forms, PSI-<br />

6130-TP and RO2433-TP, it is enticing to speculate<br />

that the two structurally different active metabolites<br />

would further increase the genetic barrier of PSI-<br />

6130.<br />

*For research use only. Not for use in diagnostic<br />

procedures.<br />

RESULTS: More than 99% of the sequences analyzed<br />

were wild type at position 282 (AGC and AGT). A<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 89


Abstract 84<br />

Determination of Lamivudine,<br />

Adefovir and Entecavir Resistance<br />

in Acute and Chronic Hepatitis B<br />

Virus Infections<br />

C Eroglu 1 , H Leblebicioglu 1 , and Members of Hepatitis<br />

Study Group 2<br />

1 Department of Infectious Diseases and Clinical<br />

Microbiology, Ondokuz Mayis University Medical School,<br />

Samsun, Turkey; 2 Hepatitis Study Group: Turkyilmaz<br />

R, Tutuncu E (Ankara), Sirmatel F (Gaziantep), Koksal<br />

I, Caylan R (Trabzon), Ozgenc O, Inan N (Izmir), Ayaz C<br />

(Diyarbakir), Aribas E (Konya), Sunbul M, Esen S, Bayirli<br />

D (Samsun), Aygen B, Yildiz O (Kayseri), Usluer G, Kartal<br />

ED (Eskisehir), Parlak M, Parlak E (Erzurum), Irmak H,<br />

Evirgen O (Van), Tulek N, Yetkin MA (Ankara), Ozsoy MF,<br />

Oncul O (Istanbul), Akbulut A, Cihangiroglu M (Elazig),<br />

Dokmetas I, Bakir M (Sivas), Ersoz G, Kaya A (Mersin),<br />

Bektas A (Samsun), Sencan I (Duzce), Akcam Z, Yayli G<br />

(Isparta), Saltoglu N, Tasova Y (Adana), Yamazhan T,<br />

Ulusoy S (Izmir), Ersoy Y (Malatya), Kilic D, Kaygusuz S<br />

(Kirikkale) Turkey<br />

The most widely used antivirals for treatment of<br />

hepatitis B virus (HBV) infection is lamivudine and<br />

adefovir. Reduced virologic response to therapy with<br />

nucleos(t)ide analogues can occur due to the emergence<br />

of mutation. Mutations conferring resistance to<br />

lamivudine, adefovir, and entecavir occur particularly<br />

in the polymerase gene. Importance of antiviral<br />

resistance to nucleos(t)ide analogues in the naïve<br />

HBV infected patients is not known. In this study, the<br />

reverse transcriptase (rt) region of polymerase gene<br />

has been sequenced to detect antiviral resistance.<br />

263 serum samples of patients with acute (n:158) and<br />

chronic (n:165) HBV infection have been investigated<br />

to detect antiviral resistance to lamivudine, adefovir,<br />

and entecavir. Mutations at the codons rtM204,<br />

rtL180, and rtV173 for lamivudine resistance, codons<br />

rtN236, rtI233, and rtA181 for adefovir resistance,<br />

and codons rtS202, rtT184, and rtI169 for entecavir<br />

resistance have been examined.<br />

Sequences have been obtained in 223 samples (118<br />

samples from acute and 105 samples from chronic<br />

HBV infection patients. Resistance for lamivudine in<br />

codon V173L has been revealed only in one case. No<br />

resistance to lamivudine at codons 180 and 204, or to<br />

adefovir at codons 181, 233, and 236, or to entecavir<br />

at codon 169, 184, and 202 have been detected.<br />

This study demonstrated the presence of mostly wild<br />

type strains in patients with acute and chronic HBV<br />

infection. It has been concluded that lamivudine,<br />

adefovir, and entecavir resistance is not an important<br />

problem in acute and chronic naïve HBV infected<br />

patients.<br />

Abstract 85<br />

Databases for Antiviral Resistance<br />

Monitoring in Hepatitis B<br />

L Yuen and S Locarnini<br />

VIDRL, North Melbourne and Evivar Medical Pty Ltd,<br />

East Melbourne, Victoria, Australia<br />

Mutations associated with antiviral drug resistance<br />

to the nucleos(t)ide analogues (NA) lamivudine<br />

(LMV), adefovir (ADV), telbivudine(LdT), tenofovir<br />

(TFV) and entecavir (ETV) have been identified for<br />

hepatitis B virus (HBV) using the SeqHepB system.<br />

SeqHepB is composed of a HBV genome sequence<br />

analysis program and a relational database (Yuen,<br />

L. et al 2007. AVR;75:64) which can then be used to<br />

correlate large numbers of patient clinical, routine<br />

pathology diagnostic data, viral mutational sequence<br />

information, and in vitro antiviral sensitivity and<br />

cross-resistance phenotypic data in an integrated and<br />

structured way for subsequent patient monitoring.<br />

The SeqHepB database currently contains routine<br />

pathology and specialised virology data for 2,004<br />

patients. Associated with these patients, there are<br />

340 clinical histories, 1,863 treatment histories,<br />

189 biopsy results, and 24,223 specimen records. In<br />

terms of routine pathology tests performed on the<br />

90 Global Antiviral Journal Volume 3, Supplement 2


samples, there are 26,029 records in the database,<br />

and these include HBV, hepatitis C virus (HCV), and<br />

hepatitis D virus (HDV) related pathology test results,<br />

as well as routine liver function and haematology<br />

test results. Samples within the database are also<br />

associated with 4,139 HBV genomic sequence<br />

information corresponding to 129,405 nucleotide or<br />

amino acid variation data points. The mutation data<br />

is correlated to an extensive data set of in-house as<br />

well as published in vitro phenotypic data on HBV<br />

antiviral drug sensitivity and resistance.<br />

The correlation of clinical, pathological and viral<br />

molecular biological data using different artificial<br />

intelligence techniques is facilitating the analysis<br />

of the pathogenesis and natural history of chronic<br />

hepatitis B in the era of antiviral drug resistance.<br />

The initial correlation of these multi-disciplinary<br />

data has identified novel mutations associated with<br />

antiviral drug resistance and cross-resistance to LMV,<br />

ADV, LdT and ETV. A linkage that exists between a<br />

3-dimensional (3-D) structure viewing program and<br />

the database enables further analysis of potentially<br />

relevant mutations within the 3D model of the<br />

polymerase.<br />

The overlap of the envelope gene with the HBV<br />

polymerase means that antiviral drug resistant<br />

HBV may have an altered envelope and that this<br />

may have public health implications. Studies<br />

have shown that lamivudine resistant HBV<br />

(rtV173L+rtL180M+rtM204V) has a significantly<br />

reduced anti-HBs binding due to important changes<br />

in the overlapping hepatitis B surface antigen<br />

(sE164D+sI195M).<br />

Chronic hepatitis B is a disease with a complex natural<br />

history, and this complexity increases with the use of<br />

antiviral agents. The SeqHepB system is an important<br />

tool that will enable the physician to individualize<br />

patient management, to cope with the explosion<br />

of antiviral associated HBV mutations, and should<br />

prove to be a useful therapeutic guide in the clinical<br />

setting as new antiviral agents and combination<br />

thereof are trialled and implemented (Shaw, T. et al<br />

2006. J.Hep;44:593).<br />

Abstract 86<br />

Combination Chemotherapy<br />

with Nucleos(t)ide Analogue<br />

Reverse Transcriptase Inhibitors<br />

(NRTI) May Suppress Multidrugresistant<br />

HBV: Using In Vitro<br />

Assays to Identify Optimal NRTI<br />

Combinations and Doses<br />

T Shaw, T Sozzi, and S Locarnini<br />

VIDRL, North Melbourne, Victoria, Australia<br />

BACKGROUND: Prolonged treatment of chronic<br />

hepatitis B (CHB) nucleos(t)ide analogue reverse<br />

transcriptase inhibitors (NRTI) almost invariably<br />

engenders viral resistance and sequential NRTI<br />

monotherapy can promote multi-drug resistance.<br />

Despite the increasing incidence of drug-resistant<br />

HBV (which can cause life-threatening complications)<br />

and previous success with combination therapy<br />

for other chronic viral infections such as HIV<br />

infection, sequential monotherapy remains the<br />

favored treatment for CHB. NRTI that have different<br />

structures and which select for different spectra of<br />

mutations are now available, suggesting that decreases<br />

in the incidence of resistance is possible by design of<br />

combination therapy optimised to individual patients<br />

and HBV phenotypes.<br />

We decided to test these postulates using a wild-type<br />

HBV and a clone generated to mimic a drug resistant<br />

clinical isolate from a patient with CHB who responded<br />

poorly to sequential treatments with adefovir (ADV),<br />

lamivudine (LMV) and entecavir (ETV). The drug<br />

resistant isolate encoded multiple substitutions in<br />

the polymerase region including rtA181T, rtI233V,<br />

rtN236T and rtM250L. (See accompanying abstract<br />

by Ayres et al.: Selection of mutlidrug resistant<br />

hepatitis B virus following sequential monotherapy<br />

and combination therapy.) As well as exhibiting<br />

a severe replication deficiency relative to WT, it<br />

showed high level resistance to L-nucleosides and<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 91


deoxyguanosine analogues and lower level resistance<br />

to adefovir and tenofovir. (See accompanying abstract<br />

by Sozzi et al.: Relative replication fitness and antiviral<br />

cross-resistance phenotypes of multidrug-resistant<br />

hepatitis b virus mutants implicated in non-response<br />

to sequential treatment with adefovir, lamivudine<br />

and entecavir.)<br />

AIMS: To compare the antiviral effects of ETV,<br />

ADV and tenofovir TFV alone and in combination<br />

replication of WT HBV and the multi-drug resistant<br />

rtA181T/I233V/N236T/ M250L mutant, as a basis for<br />

developing optimal antiviral combination therapies.<br />

METHODS: The inhibitory effects of ETV, ADV and<br />

TFV alone and in combination on in vitro replication<br />

of WT HBV and its multidrug resistant mutant<br />

derivative were compared after transfection of Huh-7<br />

cells.<br />

RESULTS: Based on EC 50<br />

ratios, the multi-drug<br />

resistant mutant displayed approximately 5, 10 and<br />

20-fold increases in resistance to ADV, TFV and ETV<br />

respectively. In general, paired drug combinations<br />

were additive or synergistic at low concentrations<br />

but antagonistic at high concentrations, presumably<br />

reflecting competition between ADV and TFV<br />

monophosphates and ETV-diphosphate for the same<br />

anabolic pathways.<br />

CONCLUSIONS: An increasing and consistent body<br />

of data to support the notion that combination<br />

therapy can decrease antiviral resistance, the<br />

endpoint likely to be of greatest long-term importance<br />

in assessing the success of treatment. In vitro<br />

phenotyping and antiviral assays should be useful<br />

for development of optimal, individually tailored<br />

combination therapy. It is likely to be more effective<br />

using combination therapy for treatment-naïve<br />

patients, rather than adding or replacing an antiviral<br />

agent after resistance develops.<br />

Abstract 87<br />

Relative Replication Capacity<br />

and Antiviral Cross-Resistance<br />

Phenotypes of Multidrugresistant<br />

Hepatitis B Virus<br />

Mutants Implicated in Nonresponse<br />

to Sequential Treatment<br />

with Adefovir, Lamivudine and<br />

Entecavir<br />

T Sozzi 1 , T Shaw 1 , FY Wong 1 , and S Locarnini 1,2<br />

1 VIDRL, North Melbourne, Victoria, Australia; 2 Evivar<br />

Medical, East Melbourne, Australia<br />

BACKGROUND: Prolonged treatment of chronic<br />

hepatitis B (CHB) nucleos(t)ide analogue reverse<br />

transcriptase inhibitors (NRTI) almost invariably<br />

engenders viral resistance. Sequencing of isolates<br />

from a patient with CHB who responded poorly to<br />

sequential treatment with adefovir, lamivudine,<br />

and entecavir revealed multiple substitutions in<br />

the polymerase region including rtA181T, rtI233V,<br />

rtN236T and rtM250L. Isolates were genotype D and<br />

HBeAg negative due to the presence of the precore [pc]<br />

G1896A mutation. All mutations were present on the<br />

same genome. (See accompanying abstract by Ayres et<br />

al.: Selection of mutlidrug resistant hepatitis B virus<br />

following sequential monotherapy and combination<br />

therapy.)<br />

AIMS: To determine antiviral drug resistance<br />

phenotype of the putative multi-drug resistant<br />

rtA181T/I233V/N236T/M250L mutant, and to<br />

determine how individual amino acid substations<br />

influence HBV replication capacity and antiviral drug<br />

resistance phenotypes.<br />

METHODS: Mutant HBV clones encoding rtA181T/<br />

N236T/M250L and rtA181T/I233V/N236T/M250L,<br />

as well as clones encoding the individual amino<br />

acid substitutions in isolation were generated by<br />

site-directed mutagenesis from a wild type (WT)


genotype D HBV isolate. WT and derived clones<br />

harboured the pc mutation, which may increase<br />

replication efficiency but does not significantly affect<br />

drug resistance. The phenotypes were determined by<br />

comparing viral replication in transfected Huh-7 cells.<br />

Virus replication and its inhibition were monitored<br />

by measuring changes in the amounts of intracellular<br />

core-associated viral DNA in cell lysates using a<br />

commercial assay kit (Seimans Versant HBV 3.0).<br />

RESULTS: The putative multi-drug resistant mutant<br />

rtA181T/I233V/N236T/M250L showed high level<br />

resistance to all four L-nucleosides tested (lamivudine,<br />

telbivudine, clevudine and emtricitabine), as well as<br />

high level resistance to the deoxyguanosine analogues<br />

entecavir, diaminopurine dioxolane, 2’,3’-dideoxy-<br />

3’-fluoroguanosine and abacavir. It also displayed<br />

lower level, but still significant, resistance to adefovir<br />

and tenofovir and a severe replication deficiency<br />

relative to WT. Except for rtA181T and rtN236T,<br />

none of the individual amino acid substitutions in<br />

isolation conferred significant drug resistance and<br />

all significantly reduced replication capacity in the<br />

absence of antiviral selection pressure. Their role(s)<br />

in antiviral resistance deserves further investigation:<br />

under antiviral selection pressure, they presumably<br />

compensate for the replication defects associated<br />

with acquisition of drug resistance and/or confer<br />

some other advantage for survival.<br />

CONCLUSIONS: Sequential treatment with NRTI<br />

promotes multi-drug resistance. Development of more<br />

rational and efficacious anti-HBV therapy will require<br />

routine genotyping and phenotypes of individual<br />

clinical isolates. It is likely that development of multidrug<br />

resistance could be reduced by combination<br />

antiviral therapy tailored to suit individual cases.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 93


Co-infection with HIV<br />

and Other Viruses<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 95


Abstract 88<br />

Treatment of HBV/HIV<br />

Co-infections<br />

MG Peters<br />

University of California, San Francisco, CA, USA<br />

Hepatitis B virus (HBV) infects an estimated 400<br />

million people worldwide, with 1 million deaths<br />

occurring annually. Transmission occurs perinatally<br />

and in childhood in countries of high endemicity<br />

but predominantly via injection drug use and sexual<br />

activity in the United States and other countries<br />

where HBV endemicity is relatively low. Given the<br />

shared modes of transmission, co-infection with HIV<br />

is common: 5%-10% of HIV-infected patients in the<br />

United States are co-infected with HBV. Infection with<br />

HDV in the US occurs in IDU. Goals of HBV therapy<br />

include sustained suppression of HBV replication;<br />

improvement in clinical liver disease; prevention of<br />

cirrhosis and subsequent liver failure; and prevention<br />

of hepatocellular carcinoma. Clearance of virus with<br />

eradication of cccDNA is not yet achievable. Clinical<br />

endpoints associated with successful therapy include<br />

loss of HBV DNA, loss of HBeAg and HBsAg, HBeAb<br />

and HBsAb seroconversion, with improvement in<br />

serum aminotransferases and liver histology.<br />

The optimal timing of HBV therapy initiation<br />

has been changing in patients with HBV/HIV coinfection<br />

due to the limited choices of HBV agents<br />

that are active against YMDD mutant virus and lack<br />

HIV activity. Treatment is indicated in those with<br />

elevated HBV viral load, ALT and the presence of<br />

fibrosis on liver biopsy. In HIV co-infected subjects,<br />

treatment depends on the need for HIV therapy. In<br />

those requiring HIV treatment, dual agents against<br />

HBV should be also included in the drug regimen.<br />

If HBV requires treatment, then consideration of<br />

early initiation of HIV should be entertained. If<br />

HIV therapy is not warranted, then options are very<br />

limited unless the patient is a candidate for interferon<br />

therapy. Adefovir may be considered, but presents<br />

a theoretical risk of development of HIV resistance<br />

to tenofovir. Telbivudine has not been studied in<br />

HBV HIV co-infected patients not receiving HAART.<br />

The following agents should be avoided without<br />

HAART: FTC, 3TC, tenofovir and entecavir as HIV<br />

resistance may emerge. Discontinuation of HIV/HBV<br />

therapy may lead to a flare in HBV with resultant<br />

liver damage. Initiation of HAART may be associated<br />

with flares due to immune reconstitution. Therefore<br />

when changing antiretroviral therapy or stopping<br />

HBV-active antiretrovirals liver function should be<br />

carefully monitored.<br />

Abstract 89<br />

Treatment of HCV in HIV-infected<br />

Individuals<br />

Z Temesgen<br />

Mayo Clinic, USA<br />

Worldwide, an estimated 40-45 million people are<br />

living with HIV/AIDS and approximately 170 million<br />

people are living with hepatitis C virus (HCV) infection.<br />

It is estimated that 25% to 30% of all HIV-infected<br />

patients also have HCV infection. Subsequent to the<br />

significant decrease in HIV-related morbidity and<br />

mortality due to highly active antiretroviral therapy<br />

(HAART), HCV-related liver disease has become a<br />

leading cause of illness and death in HIV-infected<br />

patients. While the impact of HCV on HIV is not well<br />

defined, HIV affects the natural history of HCV in a<br />

number of ways. HIV-HCV co-infected patients have<br />

higher HCV RNA levels; Progression to cirrhosis is<br />

faster in patients co-infected with HCV and HIV than<br />

those with HCV alone; Chronic HCV is a risk factor<br />

for drug-induced liver injury. Thus the treatment of<br />

HCV in patients co-infected with HIV has become<br />

important.<br />

Pegylated interferon plus ribavirin have been shown<br />

to be significantly more effective than standard<br />

interferon plus ribavirin. Sustained virologic<br />

response (SVR), defined as an undetectable HCV<br />

RNA level 6 months after the end of treatment, is<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 97


the primary goal of therapy. The secondary goals of<br />

therapy are reducing fibrosis and necroinflammation.<br />

Virologic response rates are in general lower in coinfected<br />

patients compares to responses observed<br />

in patients infected with HCV alone; only 27-40%<br />

achieving an SVR compared to more than 50% SVR<br />

in monoinfected patients. These response rates could<br />

be improved by employing weight-based ribavirin<br />

dosing, using growth factors to avoid dose reductions<br />

of ribavirin or peginterferon, and prolonging the<br />

duration of therapy. Additionally, early virologic<br />

response (EVR) has utility in identifying those with<br />

a reasonable chance of sustained response and thus<br />

avoiding unnecessary toxicity in those that do not<br />

have such chances. The absence of a 2-log decline in the<br />

level of HCV RNA after 12 weeks of therapy (no EVR)<br />

has been shown to be a potent negative predictor of<br />

treatment response. Current guidelines incorporate<br />

these data and recommend treatment with pegylated<br />

interferon plus weight-based RBV for 48 weeks<br />

provided there has been an early virologic response<br />

at week 12. The optimal duration of treatment in<br />

general as well as the issue of different durations of<br />

treatment for different subgroups of HIV and HCV<br />

co-infected patients remain to be fully defined.<br />

Abstract 90<br />

HCV/HIV Co-infections:<br />

Challenges from the Perspective of<br />

TAG<br />

T Swan<br />

Treatment Action Group (TAG), USA<br />

In the late 1980s, people with HIV/AIDS and<br />

treatment activists demanded the opportunity to<br />

participate as full partners in designing public and<br />

privately funded clinical trials of treatments for<br />

HIV and its complications. After years of struggle,<br />

they became vital participants in all stages of HIV<br />

drug development, serving on scientific peer-review<br />

committees, protocol teams, regulatory review<br />

bodies, domestic and international treatment<br />

guidelines panels; meeting regularly with sponsors of<br />

public and private research efforts; and participating<br />

in clinical trials review and data monitoring<br />

committees.<br />

By 1996, scientific innovation, diligent research,<br />

and increased research funding - due in part to<br />

activist involvement - yielded the therapeutic breakthrough<br />

known as highly-active antiretroviral therapy<br />

(HAART). HAART has dramatically improved the<br />

prognosis for HIV, saving at least three million years<br />

of life in the US alone.<br />

Now, hepatitis C co-infection has emerged as a<br />

significant threat to health, quality of life, and survival<br />

of HIV positive people, among whom it is prevalent.<br />

Globally, four to five million people are HIV/HCV<br />

co-infected. In the US, approximately 30% of HIVpositive<br />

people are co-infected, and more than 45%<br />

in Southern and Eastern Europe.<br />

End-stage liver disease from hepatitis C is now a<br />

leading cause of death among HIV-positive people<br />

in the US and Europe. HIV accelerates hepatitis<br />

C progression, doubles the risk for cirrhosis, and<br />

shortens survival after hepatic decompensation.<br />

The current standard of care for HCV fails to eradicate<br />

the virus in the majority of co-infected people. Side<br />

effects are debilitating, sometimes life-threatening.<br />

Many are reluctant to undergo treatment, or cannot<br />

complete it.<br />

AIDS treatment activists have sharpened their focus<br />

on the urgent need for efficient hepatitis C research<br />

and drug development. The spectacular successes,<br />

and challenges in HIV drug development and clinical<br />

care offer important lessons for HCV.<br />

Effective combination therapy, and adherence<br />

support programs will prevent emergence of drug<br />

resistance. Sponsors should collaborate to support<br />

multi-experimental agent trials, as has been done<br />

recently in HIV research.<br />

New HCV therapies need to be studied in people<br />

with urgent unmet medical need: liver transplant<br />

98 Global Antiviral Journal Volume 3, Supplement 2


candidates and recipients, HIV/HCV co-infected<br />

people, and nonresponders. Drug-drug interaction<br />

studies should be carried out early, to facilitate these<br />

trials, and to guide real-world use.<br />

Sponsors should provide pre-approval access to<br />

experimental agents in people who need them most.<br />

Expanded access is the right thing to do; it gives<br />

people without options access to potentially lifesaving<br />

treatment; broadens the pool of physicians<br />

with experience using new treatments; and gathers<br />

safety data.<br />

Registration trials for novel HCV therapies should help<br />

to define optimal treatment strategies, side effects<br />

management, and best practices for delivering care<br />

and treatment in addition to shortening treatment<br />

duration, and increasing efficacy to increase HCV<br />

treatment access, uptake, completion, and success<br />

rates.<br />

Abstract 91<br />

Tolerability of Darunavir/<br />

Ritonavir Versus Lopinavir/<br />

Ritonavir in Lopinavir/Ritonavirnaïve,<br />

Treatment-experienced,<br />

Hepatitis B or C Co-infected<br />

Patients in TITAN<br />

D Jayaweera 1 , E De Paepe 2 , JM Mrus 3 , YK Dayaram 3 ,<br />

and F Tomaka 4<br />

1 University of Miami, Miami, FL, USA; 2 Tibotec BVBA,<br />

Mechelen, Belgium; 3 Tibotec Therapeutics, Bridgewater,<br />

NJ, USA; 4 Tibotec Inc., Yardley, PA, USA<br />

BACKGROUND: The randomized, controlled, phase<br />

III TITAN trial in HIV-1-infected lopinavir/ritonavir<br />

(LPV/r)-naïve, treatment-experienced patients<br />

showed that darunavir (DRV) with low-dose ritonavir<br />

(DRV/r) was generally well tolerated. This analysis<br />

compared the safety and tolerability of DRV/r versus<br />

LPV/r in patients with hepatitis B or C (HBV/HCV)<br />

co-infection.<br />

METHODS: Patients with HIV-1 RNA >1,000 copies/<br />

mL were on stable HAART for ≥12 weeks or offtreatment<br />

for ≥4 weeks. Patients received DRV/r<br />

600/100mg bid or LPV/r 400/100mg bid, plus 2–3<br />

optimized NRTIs/NNRTIs. Patients with chronic<br />

HBV/HCV co-infection were eligible if their condition<br />

was stable and they would not require treatment for<br />

their hepatitis; those with significantly decreased<br />

hepatic function or hepatic decompensation were<br />

excluded.<br />

RESULTS: Of the 298 and 297 patients randomized<br />

to DRV/r and LPV/r (overall baseline median CD4<br />

count = 232 cells/mm 3 ), 52 (18%) and 37 (13%),<br />

respectively, had HBV/HCV co-infection at baseline.<br />

The most common Grade 3-4 liver-related laboratory<br />

abnormalities in co-infected patients were increased<br />

alanine aminotransferase (ALT; 12% DRV/r versus<br />

25% LPV/r) and increased aspartate aminotransferase<br />

(AST; 10% DRV/r versus 22% LPV/r). The overall<br />

incidence of liver-related adverse events was higher in<br />

co-infected patients (25% DRV/r versus 24% LPV/r)<br />

than in non-co-infected patients (4% DRV/r versus<br />

4% LPV/r). The most commonly observed (>5% in<br />

co-infected patients) liver-related adverse events<br />

were increased gamma glutamyltransferase, ALT,<br />

and AST. Although clinical jaundice was reported in<br />

6% DRV/r versus 0% LPV/r, the overall incidence of<br />

hyperbilirubinemia in co-infected patients was similar<br />

in both arms. All of these liver-related adverse events<br />

were reported in


Abstract 92<br />

The Effect of Lopinavir/Ritonavir<br />

on HIV/HCV Co-Infected<br />

Individuals<br />

CA Smith 1 , P Greiger-Zanlungo 2 , K Crain 3 , B Morgan 4 ,<br />

R Stubbs 5 , and R Rode 5<br />

1 Mount Morris Medical, New York, NY, USA; 2 Mount<br />

Vernon Hospital, New York, NY, USA; 3 Penn State, PA,<br />

USA; 4 Morgan State, Baltimore, MD, USA; 5 Abbott<br />

Laboratories, Chicago, IL, USA<br />

Background: Over the past several years the<br />

Black and Hispanic communities have seen an<br />

increase in the prevalence and incidence of Human<br />

Immunodeficiency Virus (HIV) and Hepatitis C Virus<br />

(HCV) and the common side effect of liver toxicity of<br />

antiretroviral therapy. Overall, liver toxicity leads to<br />

treatment discontinuation in an estimated 10-15%<br />

of patients who initiate HAART in the United States.<br />

Our study reviewed use of antiretroviral therapy in<br />

HIV/HCV co-infected individuals and the incidence<br />

of severe liver toxicity after initiation of LPV/RIT<br />

and other highly active antiretroviral medications.<br />

Methods: A Retrospective chart review of HIV/<br />

HCV co-infected patients on Lopinavir/Ritonavir was<br />

conducted over a 12 - 18 month period of time in 2<br />

inner city hospitals. All patients were 18 years and<br />

older and tested positive for HIV-1 and Hepatitis C<br />

viruses. The following demographic information was<br />

obtained: age, ethnicity, gender, and risk factor. The<br />

following was also obtained: LFT’s, CD4, HIV & HCV<br />

viral load, lipids, and antiretroviral usage.<br />

Results: Over 120 charts were reviewed but 76<br />

charts were analyzed for this study. The mean age<br />

was 44.1 with a range from 40 to 48. 12% (9) of had<br />

history of DM; 93% (70) had no history of Alcohol<br />

use; 48% (36) were smokers. 67% (50) were Black;<br />

16% (12) were Hispanic; 13% (10) were Caucasian<br />

and 4% (3) were other. 23% were HAV positive;<br />

31% positive for HBV (20% antigen positive). The<br />

following HCV genotypes were represented: 34% 1A;<br />

24% 1B; 28% of charts did not have HCV genotype<br />

documented. Only 6% of patients had ever been<br />

treated for HCV. 45% of patients were ARV naïve and<br />

24% were ARV experienced. 44% of patients were<br />

on taking Lopinavir/Ritonavir with the predominant<br />

ART backbone being Zidovudine +Lamivudine (29%),<br />

tenofovir + Emtricitabine (12%), and Abacavir +<br />

Zidovudine + Lamivudine. 84% of patients had a<br />

undetectable HIV viral load 6 months after initiation<br />

and less than 10% of patients had ALT and AST<br />

elevations. No patients discontinued LPV/RIT due to<br />

liver toxicity.<br />

Conclusion: Lopinavir/Ritonavir was the most<br />

frequently used protease inhibitor in this inner city<br />

setting of HIV/HCV co-infected individuals. Lopinavir/<br />

Ritonavir is an effective option for the treatment of<br />

co-infected HIV/HCV infected individuals when used<br />

in combination with other antiretroviral agents.<br />

Abstract 93<br />

Pharmacokinetics of TMC125<br />

in HIV-negative Volunteers<br />

with Mild or Moderate Hepatic<br />

Impairment<br />

TN Kakuda 1 , M Schöller-Gyüre 2 , F Tomaka 1 ,<br />

G De Smedt 2 , B Woodfall 2 , C Berckmans 2 , M Peeters 2 ,<br />

and RMW Hoetelmans 2<br />

1 Tibotec Inc, Yardley, PA, USA; 2 Tibotec BVBA,<br />

Mechelen, Belgium<br />

BACKGROUND: TMC125 (etravirine; ETR) is a next<br />

generation NNRTI with potent activity against both<br />

wild-type HIV and viruses resistant to currently<br />

approved NNRTIs. TMC125 is mainly eliminated via<br />

the hepatobiliary route. This study aimed to assess<br />

the pharmacokinetics of TMC125 in HIV-negative<br />

volunteers with hepatic impairment compared to<br />

healthy controls.<br />

METHODS: An open-label trial was conducted.<br />

Volunteers with mild or moderate hepatic impairment<br />

100 Global Antiviral Journal Volume 3, Supplement 2


(Child-Pugh A or B, respectively) and healthy<br />

controls matched for age, gender, race, and BMI were<br />

included. All volunteers received TMC125 200 mg bid<br />

for 7 days with a morning dose on Day 8. TMC125<br />

pharmacokinetics over 12 hours on Days 1 and 8 were<br />

determined using non-compartmental methods and<br />

analyzed by a linear mixed effects model. Safety and<br />

tolerability were assessed.<br />

RESULTS: TMC125 pharmacokinetics in volunteers<br />

with mild (n=8, median age 57 years, 5 males) and<br />

moderate (n=8, median age 54 years, 6 males) hepatic<br />

impairment were comparable to matched healthy<br />

controls (n=16). In volunteers with mild hepatic<br />

impairment, relative to controls, the least squares<br />

means (LSM) ratios (90% confidence intervals [CI])<br />

for TMC125 exposure (AUC 12h<br />

) and maximum (C max<br />

)<br />

plasma concentration on Day 1 were 0.99 (0.75–1.29)<br />

and 0.92 (0.69–1.21), respectively. On Day 8, AUC 12h<br />

,<br />

C max<br />

, and minimum plasma concentration (C min<br />

) were<br />

0.87 (0.69–1.09), 0.79 (0.63–1.00), and 0.87 (0.65–<br />

1.17), respectively, relative to controls. For volunteers<br />

with moderate hepatic impairment, relative to<br />

controls, LSM ratios and 90% CI for AUC 12h<br />

and C max<br />

on Day 1 were 0.77 (0.55–1.08) and 0.63 (0.47–0.85),<br />

respectively. On Day 8, AUC 12h<br />

, C max<br />

, and C min<br />

were<br />

0.82 (0.60–1.11), 0.72 (0.54–0.96), and 0.98 (0.68–<br />

1.42), respectively, relative to controls. All treatment<br />

emergent adverse events were mild or moderate (Grade<br />

1 and 2) in severity. The most common adverse events<br />

for volunteers with mild hepatic impairment were<br />

fatigue and nausea (each occurring in 2 volunteers<br />

with hepatic impairment and 1 healthy volunteer).<br />

Dizziness and muscle spasms were the most common<br />

adverse events for volunteers with moderate liver<br />

impairment (each occurring in 2 volunteers with<br />

hepatic impairment and none in healthy volunteers).<br />

Headache was reported in 4 healthy volunteers, but<br />

not in volunteers with liver impairment. No rash was<br />

reported. There were no clinically significant changes<br />

in laboratory (including hepatic) or cardiovascular<br />

safety parameters.<br />

CONCLUSIONS: The systemic exposure to TMC125 in<br />

volunteers with hepatic impairment was comparable<br />

to that in healthy subjects. TMC125 was generally safe<br />

and well tolerated. No dose adjustments of TMC125<br />

are necessary in patients with mild or moderate<br />

hepatic impairment.<br />

Abstract 94<br />

Tolerability of Once-daily<br />

Darunavir/r versus Lopinavir/r<br />

in Treatment-naïve, Patients<br />

Co-infected with Hepatitis B<br />

and/or C in the ARTEMIS Trial<br />

R Ortiz 1 , J Fourie 2 , J Andrade-Villanueva 3 , S Dincq 4 ,<br />

C Vanden Abeele 4 , F Tomaka 5 , and L Lavreys 4<br />

1 Orlando Immunology Center, Orlando, USA; 2 Chronic<br />

Diseases of Lifestyle Unit, Tygerberg, South Africa;<br />

3 Hospital Civil de Guadalajara, Guadalajara, Mexico;<br />

4 Tibotec BVBA, Mechelen, Belgium; 5 Tibotec Inc,<br />

Yardley, PA, USA<br />

BACKGROUND: In the randomized, controlled, Phase<br />

III ARTEMIS trial in HIV-1-infected treatment-naïve<br />

patients, once-daily darunavir/r (DRV/r 800/100mg)<br />

was generally well tolerated and had a lower incidence<br />

of treatment-related, moderate-to-severe diarrhea<br />

and triglyceride elevations versus lopinavir/r (LPV/r).<br />

For treatment-experienced patients, including those<br />

co-infected with hepatitis B and/or C virus (HBV/<br />

HCV), DRV/r 600/100 mg bid has been shown to<br />

be a well-tolerated treatment option. This analysis<br />

examined the safety and tolerability of once-daily<br />

DRV/r (800/100mg) and LPV/r (800/200 mg total<br />

daily dose) in treatment-naïve HBV/HCV co-infected<br />

ARTEMIS patients.<br />

METHODS: Patients with HIV-1 RNA ≥5,000<br />

copies/mL received DRV/r 800/100mg qd or LPV/r<br />

800/200mg total daily dose, plus a fixed-dose<br />

combination tablet of tenofovir (300mg qd) and<br />

emtricitabine (200mg qd). Patients with chronic<br />

HBV/HCV co-infection were permitted to enter the<br />

trial if their condition was clinically stable and they<br />

would not require treatment for their hepatitis. HBV<br />

was determined by HB surface antigen testing and<br />

HCV by HCV antibody and PCR testing.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 101


RESULTS: 343 and 346 patients were randomized to<br />

DRV/r and LPV/r, respectively; 43 (13%) of DRV/r<br />

and 48 (14%) of LPV/r patients had HBV/HCV coinfection.<br />

The overall incidence of liver-related adverse<br />

events (AEs) was higher in co-infected patients<br />

(7/43, 16% DRV/r; 18/48, 38% LPV/r) than in nonco-infected<br />

patients (9/300, 3% DRV/r; 13/298, 4%<br />

LPV/r). In co-infected patients, the most common<br />

events (>5%) were liver-function abnormalities<br />

reported as AEs: increased ALT, increased AST and<br />

increased transaminases. These AEs were reported<br />

in ≤1% of non-co-infected patients. There were few<br />

clinical AEs, with the exception of HCV, which was<br />

reported in three (6%) LPV/r co-infected patients; all<br />

other AEs were reported in at most one co-infected<br />

patient.<br />

Regarding laboratory abnormalities, the observed<br />

incidences of increases in ALT and AST were higher<br />

in patients with HBV/HCV co-infection compared<br />

with those without co-infection in both treatment<br />

groups. The incidence of Grade 3 increases in ALT<br />

or AST was comparable in both co-infected groups.<br />

Grade 4 increases in ALT or AST were not observed<br />

in DRV/r patients with co-infection but were seen in<br />

6/48 (13%) and 4/48 (8%) co-infected LPV/r patients,<br />

respectively. In the LPV/r group, the overall incidence<br />

of hyperbilirubinemia was higher in co-infected<br />

patients (6/48, 13%) versus non-co-infected patients<br />

(13/298, 4%), while this was low and comparable for<br />

co-infected and non-co-infected DRV/r patients (1/43<br />

and 5/300, 2%).<br />

CONCLUSIONS: Although HBV/HCV co-infected<br />

patients had a higher incidence of liver-related AEs<br />

compared with non-co-infected patients, the most<br />

commonly observed were liver-function abnormalities<br />

reported as AEs. There were no Grade 4 ALT or AST<br />

elevations in DRV/r co-infected patients, while these<br />

occurred in LPV/r patients. These results support<br />

previous findings and suggest that safety monitoring<br />

of HIV-1-infected patients with HBV/HCV coinfection<br />

treated with once-daily DRV/r 800/100mg<br />

is appropriate.<br />

Abstract 95<br />

Pharmacokinetics of Multipledose<br />

Darunavir in Combination<br />

with Low-dose Ritonavir in<br />

Individuals with Impaired Hepatic<br />

Function<br />

F Tomaka 1 , RM Hoetelmans 2 , S Spinosa-Guzman 2 ,<br />

E De Paepe 2 , T Stevens 1 , M De Pauw 2 , JM Mrus 3, and<br />

VJ Sekar 1<br />

1 Tibotec Inc., Yardley, PA, USA; 2 Tibotec BVBA,<br />

Mechelen, Belgium; 3 Tibotec Therapeutics, Bridgewater,<br />

NJ, USA<br />

BACKGROUND: Darunavir (DRV), an HIV protease<br />

inhibitor potent against wild-type and protease<br />

inhibitor-resistant HIV, is co-administered with lowdose<br />

ritonavir (RTV; DRV/r). This study assessed the<br />

pharmacokinetics and safety of DRV/r 600/100mg<br />

bid in HIV-negative volunteers with mild or moderate<br />

liver impairment compared with matched HIVnegative,<br />

healthy controls.<br />

METHODS: Liver impairment was defined according<br />

to Child-Pugh classification A (mild) and B (moderate).<br />

Volunteers received DRV/r 600/100mg bid for 6<br />

days with a morning dose on Day 7. Volunteers also<br />

received RTV 100mg on the evening of Day 7 and<br />

in the morning and evening of Days 8 and 9. Full<br />

pharmacokinetic profiles were obtained up to 72<br />

hours post-dose for DRV and 12 hours post-dose<br />

for RTV on Day 7. Safety and tolerability were also<br />

assessed.<br />

RESULTS: DRV pharmacokinetics in volunteers with<br />

mild (n=8) or moderate (n=8) liver impairment were<br />

comparable to healthy controls (n=8 in each group).<br />

In those with mild liver impairment, relative to<br />

controls, the least squares means (LSM) ratios (90%<br />

confidence intervals [CI]) for DRV exposure (AUC 12h<br />

),<br />

maximum (C max<br />

), and minimum (C min<br />

) plasma<br />

concentrations at Day 7 were 0.94 (0.75–1.17), 0.88<br />

(0.73–1.07), and 0.83 (0.63–1.10), respectively.<br />

102 Global Antiviral Journal Volume 3, Supplement 2


For those with moderate liver impairment, LSM<br />

ratios and 90% CI for AUC 12h<br />

, C max<br />

, and C min<br />

at Day<br />

7 were 1.20 (0.90–1.60), 1.22 (0.95–1.56), and 1.27<br />

(0.87–1.85), respectively, relative to controls. DRV/r<br />

treatment was generally well tolerated, regardless of<br />

degree of liver impairment level. The most commonly<br />

reported adverse events during DRV/r treatment<br />

in volunteers with mild liver impairment were<br />

increased alanine aminotransferase, increased<br />

aspartate aminotransferase, and headache, each<br />

reported in two volunteers; these were also each<br />

reported in two healthy volunteers. The most<br />

commonly reported adverse event during DRV/r<br />

treatment in volunteers with moderate liver<br />

impairment was fatigue, reported in 2 volunteers; and<br />

in none of the healthy volunteers. All adverse events<br />

were Grade 1–2 in severity except for one Grade 3<br />

increase in alanine aminotransferase in a subject with<br />

mild liver impairment. No consistent or clinically<br />

relevant changes in laboratory parameters were<br />

observed. No adverse events led to discontinuation<br />

and no serious adverse events were reported.<br />

CONCLUSIONS: DRV pharmacokinetics in subjects<br />

with mild or moderate liver impairment were<br />

comparable to healthy controls. No serious adverse<br />

events, or adverse events leading to discontinuation,<br />

were reported. For patients with mild or moderate liver<br />

impairment, no dose adjustments are necessary and<br />

routine clinical monitoring is considered adequate.<br />

Abstract 96<br />

TMC125 Safety and Tolerability in<br />

Treatment-Experienced Hepatitis<br />

B or C Co-infected Patients in<br />

DUET-1 and DUET-2<br />

T Campbell 1 , A Mills 2 , P Morlat 3 , M Schechter 4 ,<br />

G De Smedt 5 , M Peeters 5 , F Tomaka 6 , and B Woodfall 5<br />

1 University of Colorado Health Sciences Center, Denver,<br />

USA; 2 Private Practice, Los Angeles, USA; 3 Saint Andre<br />

Hospital, University Victor Segalen, Bordeaux, France;<br />

4 Universidade Federal do Rio de Janeiro, Brazil; 5 Tibotec<br />

BVBA, Mechelen, Belgium; 6 Tibotec Inc, Yardley, PA, USA<br />

Background: DUET-1 and DUET-2 are ongoing,<br />

randomized, double-blind, placebo-controlled<br />

trials of identical design which investigate TMC125<br />

(etravirine; ETR) in treatment-experienced, HIV-1-<br />

infected patients. We report safety results by baseline<br />

hepatitis co-infection status from a planned 24-week<br />

pooled analysis.<br />

Methods: Patients with documented (historical<br />

data and/or viral genotype) NNRTI resistance and<br />

with ≥ 3 primary PI mutations on stable virologically<br />

failing treatment were randomized to TMC125 200mg<br />

or placebo bid, each administered with darunavir/<br />

ritonavir, optimized NRTIs +/– enfuvirtide. To be<br />

eligible, patients co-infected with chronic hepatitis<br />

B or C (HBV/HCV) had to be clinically stable with<br />

AST/ALT


serious hepatic AEs and hepatic AEs leading to<br />

discontinuation among co-infected subjects was<br />

comparable between the treatment groups. In both<br />

treatment groups, grade 3/4 AST/ALT elevations<br />

were more frequent in those who were co-infected;<br />

differences between TMC125 and placebo were<br />

small.<br />

Conclusions: Consistent with the underlying<br />

chronic hepatitis co-infection, hepatic AEs and<br />

elevated hepatic parameters were more frequent in<br />

co-infected patients compared with non-co-infected<br />

patients. The incidence and severity of hepatic AEs<br />

with TMC125 were generally similar to placebo,<br />

irrespective of hepatitis co-infection status. TMC125<br />

does not appear to increase hepatic toxicity in patients<br />

with hepatitis co-infection.<br />

104 Global Antiviral Journal Volume 3, Supplement 2


Pharmacology and<br />

Drug Interactions of HBV<br />

and HCV Therapeutics<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 105


ABSTRACT 97<br />

Preclinical Drug Metabolism in<br />

Viral Hepatitis Drug Discovery<br />

AS Ray<br />

Department of Drug Metabolism, Gilead Sciences, Inc.,<br />

California, USA<br />

Approximately 350 and 170 million people worldwide<br />

are chronically infected by the hepatitis B and C<br />

viruses (HBV and HCV), respectively. Identification of<br />

more effective therapies for these viruses will greatly<br />

reduce morbidity and mortality due to liver disease.<br />

Potent inhibitors of the HCV NS3/4A protease and<br />

nucleoside and non-nucleoside inhibitors of the<br />

NS5B RNA dependent RNA polymerase (RdRp) have<br />

been identified in vitro. Use of metabolism assays<br />

and pharmacokinetic studies in animals will allow for<br />

some of these compounds to bridge the gap between<br />

the laboratory and the clinic, eventually redefining<br />

HCV therapy. Targeting viral replication in the liver<br />

presents many unique opportunities and challenges.<br />

The direct flow of portal blood to the liver and the<br />

expression of multiple influx transporters results in<br />

the potential for concentration of antiviral agents<br />

in the liver. However, the liver has a high capacity to<br />

convert xenobiotics into inactive metabolites through<br />

oxidation and conjugation and to reduce intrahepatic<br />

levels through efflux transport. The unique<br />

requirement for phosphorylation of nucleoside and<br />

nucleotide inhibitors makes the assessment of liver<br />

levels of the active nucleoside triphosphate analog<br />

of paramount importance in predicting efficacy.<br />

Nucleosides and nucleotides are effective therapies<br />

for HBV and a number of potent agents have either<br />

recently been approved by regulatory agencies or<br />

are in late stages of clinical development. Similarly,<br />

nucleoside inhibitors of the HCV RdRp promise<br />

to be an important part of future virally targeted<br />

combination therapy for HCV. Non-nucleoside and<br />

protease inhibitor levels in the liver can be more closely<br />

estimated based on their non-protein bound plasma<br />

concentrations by assessment of individual rates of<br />

liver accumulation, catabolism and excretion. Various<br />

strategies for predicting human pharmacokinetics<br />

and, ultimately, clinical efficacy will be discussed,<br />

highlighting examples from data generated with HBV<br />

and HCV inhibitors.<br />

Abstract 98<br />

Frontiers of Clinical Pharmacology<br />

in Hepatitis Drug Development<br />

CW Flexner<br />

Pharmacology and Molecular Sciences, and International<br />

Health, Johns Hopkins University, Baltimore, MD, USA<br />

As the number of drugs approved for the treatment<br />

of HBV and HCV infection continues to grow,<br />

the pharmacologic consequences of combination<br />

chemotherapy, including drug interactions, will become<br />

more complex. Principles of clinical pharmacology<br />

have informed the rational development of treatment<br />

regimens for HIV, and many of these principles can be<br />

applied to hepatitis virus infections.<br />

There are currently 24 antiretroviral drugs approved<br />

for use in the United States; three of these –<br />

lamivudine, emtricitabine, and tenofovir – also have<br />

activity against HBV. In addition, three drugs approved<br />

for the treatment of HBV or HCV – interferon-α,<br />

adefovir, and entecavir – have anti-HIV activity.<br />

Clearly our lives — as care providers, as patients,<br />

as citizens concerned about these epidemics — are<br />

getting more complicated. The relationship between<br />

drug concentrations, antiviral activity, toxicity, and<br />

resistance is a complex function within any individual<br />

patient. The makeup of a multidrug regimen, drug<br />

tolerance, adherence patterns, and sensitivity of the<br />

virus to individual drugs in the regimen all contribute<br />

to treatment success or failure. The availability of<br />

an increased number of agents makes possible the<br />

design of multi-drug regimens with activity against<br />

multiple viruses, and these regimens may be used in<br />

the clinic before they can be adequately evaluated in<br />

clinical trials. As a consequence, surprises can -- and<br />

will -- occur.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 107


Although there is an increasing interest in the<br />

influence of individual genetic make-up on<br />

drug concentrations and drug response (i.e.,<br />

pharmacogenetics), environmental factors such as<br />

food effects, patient adherence behavior, interactions<br />

with concomitant prescription drugs, concomitant<br />

herbal medicines, and concomitant disease can also<br />

have a profound impact on treatment outcome.<br />

Pharmacokinetic drug interactions are becoming<br />

more numerous and more complex. The number of<br />

potential 3- and 4-drug combinations for HIV treatment<br />

alone now number in the thousands. It is therefore<br />

not possible to prospectively study all potential<br />

drug combinations before they might be used in the<br />

clinic. This is especially problematic for treatmentexperienced<br />

patients, whose regimens might be based<br />

on resistance patterns that necessitate the use of an<br />

obscure regimen. Several recent examples highlight<br />

the hazards of using untested regimens. Combination<br />

studies with standard doses of tenofovir (TDF) and<br />

didanosine (ddI) resulted in unexpectedly high<br />

didanosine concentrations, and may have produced<br />

a greater than expected frequency of didanosine<br />

toxicities (Robbins et al., 2003). Three studies in<br />

treatment naïve patients found an unacceptably high<br />

early failure rate with combinations of tenofovir,<br />

didanosine, and lamivudine, or tenofovir, abacavir,<br />

and lamivudine (Jemsek et al., 2004; Landman et<br />

al., 2004). In addition, a recent report highlights an<br />

unexpected association between early non-response<br />

to HCV treatment and simultaneous use of abacavir<br />

(Bani-Sadr et al., 2007). Ribavirin is an inhibitor of<br />

inosine 5′-monophosphate dehydrogenase, and is<br />

antagonistic in vitro with the antiretroviral nucleosides<br />

didanosine, zidovudine, and didanosine. A clinical<br />

pharmacological interaction between abacavir and<br />

ribavirin has been postulated but not proven.<br />

The mechanisms for these unexpected outcomes are<br />

not yet completely understood. In the case of the TDF<br />

and ddI interaction, there appears to be a previously<br />

unexpected pharmacokinetic interaction between<br />

these drugs, perhaps involving an inhibition by TDF<br />

of ddI degradation by the enzyme purine nucleoside<br />

phosphorylase (Ray et al., 2003). TDF and ddI do not<br />

interfere with intracellular phosphorylation of the<br />

other agent when both are applied to human cells<br />

(Robbins et al., 2003). In the case of the TDF-ddI<br />

and TDF-abacavir failures, the final answer is not yet<br />

known. However, these regimens may be associated<br />

with early emergence of the nucleoside resistance<br />

mutation K65R (Landman et al., 2004), resulting in<br />

an unexpectedly high rate of early genetic resistance.<br />

This may represent a pharmacodynamic, rather than<br />

a pharmacokinetic interaction.<br />

Polypharmacy is a growing complication of HIV, HCV,<br />

and HBV infections. As the number of agents effective<br />

in the management of these infections continues<br />

to grow, patients and physicians are expected to<br />

deal with the increasingly complex potential for<br />

pharmacokinetic and pharmacodynamic drug<br />

interactions (Piscitelli and Gallicano, 2001). The HIV<br />

protease inhibitors and the nonnucleoside reverse<br />

transcriptase inhibitors are especially susceptible<br />

to pharmacokinetic drug-drug interactions. The<br />

potent inhibition of CYP 3A4 by ritonavir produced<br />

the unexpected finding that ritonavir increased the<br />

AUC and Cmax of fluticasone, when administered<br />

as a nasal spray, by approximately 350-fold and 25-<br />

fold respectively. This significant increase in plasma<br />

fluticasone resulted in a significant decrease (86%)<br />

in plasma cortisol. This has produced features of<br />

Cushing’s syndrome in some patients, and the<br />

possibility of secondary adrenal insufficiency when<br />

fluticasone is stopped (Andrade and Flexner, 2000).<br />

A better understanding of mechanisms of drug<br />

interaction should lead us in the future to improved<br />

strategies for managing complex regimens in our<br />

patients.<br />

Suggested Reading:<br />

Andrade A, Flexner C. HIV-related drug metabolism<br />

and cytochrome P450 enzymes. AIDS Clin Care.<br />

2000;12:91-95.<br />

Bani-Sadr F, Denoeud L, Morand P, Lunel-Fabiani F,<br />

Pol S, Cacoub P, Perronne C, Carrat F; Agence Nationale<br />

pour la Recherche contre le SIDA et les Hepatites<br />

Virales HC02-Ribavic Study Team. Early virologic<br />

failure in HIV-co-infected hepatitis C patients treated<br />

108 Global Antiviral Journal Volume 3, Supplement 2


with the peginterferon-ribavirin combination: does<br />

abacavir play a role? J Acquir Immune Defic Syndr.<br />

2007;45:123-5.<br />

Flexner C. HIV drug development: the next 25 years.<br />

Nature Reviews Drug Discovery 2007; in press. Online<br />

version published October 12, 2007.<br />

Jemsek J, Hutcherson P, Harper E. Poor virologic<br />

responses and early emergence of resistance in<br />

treatment naïve, HIV-infected patients receiving a<br />

once daily regimen of didanosine, lamivudine, and<br />

tenofovir DF. In Program and Abstracts of the Eleventh<br />

Conference on Retroviruses and Opportunistic<br />

Infections. San Francisco, CA, February, 2004.<br />

Landman R, Peytavin G, Descamps D, et al. Low<br />

genetic barrier to resistance is a possible cause of<br />

early virologic failures in once-daily regimen of<br />

abacavir, lamivudine, and tenofovir: the Tonus Study<br />

. In Program and Abstracts of the Eleventh Conference<br />

on Retroviruses and Opportunistic Infections. San<br />

Francisco, CA, February, 2004.<br />

Nettles RE, Kieffer TL, Parsons T, Johnson J,<br />

Cofrancesco J, Gallant JE, , Carson K, Siliciano RF,<br />

Flexner C. Marked intraindividual variability in<br />

antiretroviral concentrations may limit the utility of<br />

therapeutic drug monitoring. Clin Infect Dis 2006; 42:<br />

1189-1196.<br />

Piscitelli SC, Gallicano KD. Interactions among drugs<br />

for HIV and opportunistic infections (review). N Engl<br />

J Med. 2001;34:984-96.<br />

Ray AS, Mahmoudi A, Fridland A. Mechanism of 2’,<br />

3’-dideoxyinosine’s drug interaction with allopurinol,<br />

ganciclovir, and tenofovir disoproxil fumarate<br />

(abstract). Antiviral Res. 2003;57:A50.<br />

Robbins BL, Wilcox CK, Fridland A, Rodman JH.<br />

Metabolism of tenofovir and didanosine in quiescent<br />

or stimulated human peripheral blood mononuclear<br />

cells. Pharmacotherapy 2003 Jun;23(6):695-701.<br />

Washington CB, Flexner C, Sheiner LB, Rosenkranz<br />

SL, Segal Y, Aberg JA, Blaschke TF, for the ACTG 378<br />

Study Team. Effect of simultaneous versus staggered<br />

dosing on pharmacokinetic interactions of protease<br />

inhibitors. Clin Pharmacol Ther 2003; 73: 406-416.<br />

Abstract 99<br />

Current and Future: HCV<br />

Maintenance Therapy<br />

KL Lindsay<br />

University of Southern California, 1640 Marengo Street,<br />

Suite 103, Los Angeles, CA 90033 USA<br />

Background: Three large maintenance peginterferon<br />

therapy trials were initiated in the late<br />

1990’s to evaluate the effectiveness of long-term<br />

low-dose peginterferon alfa treatment of patients<br />

who had failed to develop a virological response<br />

to peginterferon + ribavirin combination therapy.<br />

Patients with clinically compensated liver disease<br />

and advanced hepatic fibrosis were included,<br />

and randomized to receive either peginterferon<br />

monotherapy or no treatment in two studies or<br />

colchicine in the third study.<br />

Status of peginterferon maintenance<br />

trials: Primary study endpoint data from the EPIC-3<br />

study are anticipated in 2009. A planned interim<br />

analysis of the COPILOT study in 2005 demonstrated<br />

a reduction in portal hypertension outcomes in<br />

patients treated with peginterferon compared to<br />

those in the colchicine group. Primary outcome data<br />

is now available from the HALT-C trial, in which 1050<br />

patients who had not responded to PEG IFN + RBV<br />

therapy, with liver biopsy Ishak fibrosis scores ≥ 3<br />

were randomized to receive either peginterferon alfa-<br />

2a 90 mcg weekly (n=517) or no treatment (n=533)<br />

for 3.5 years. Clinical outcomes were monitored, and<br />

liver biopsies were repeated 1.5 and 3.5 years after<br />

randomization. For patients without cirrhosis at<br />

baseline, a ≥ 2 point increase in the fibrosis score on<br />

follow-up liver biopsies defined a histologic outcome.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 109


At the end of the study the frequency of outcomes<br />

in the treated group and the control group was not<br />

different (34.1% vs. 33.8%, hazard ratio = 1.01; 95%<br />

CI = 0.84-1.26; p=0.91). However, mean serum ALT<br />

and HCV RNA levels decreased significantly with<br />

treatment (both p


CONCLUSIONS: An HCV replicon over-expressing<br />

the MDR1 protein, Pgp, was established as a screen<br />

for monitoring whether HCV inhibitors are Pgp<br />

substrates. A positive correlation was demonstrated<br />

between the more classical caco bidirectional assay<br />

and the HCV replicon Pgp system. Advantages of<br />

employing the robust HCV replicon Pgp assay include<br />

the requirement of lower compound concentrations<br />

as well as providing a direct measure of the effect<br />

of Pgp efflux on the potency of any HCV replication<br />

inhibitor.<br />

Abstract 101<br />

Pharmacologic Evaluation of<br />

Novel Small Molecule HCV<br />

Inhibitors Affecting NS5Adependent<br />

Functions<br />

CR Wobbe, NJ Cao, H-J Cho, R Fathi, A Lam, G Li,<br />

Y Liao, K Nawoschik, A Sandrasagra, G Westby, R Wu,<br />

Z Yang, and Q Zhu<br />

XTL Biopharmaceuticals, Valley Cottage, NY, USA<br />

Background: HCV NS5A is considered to be a<br />

promising target for antiviral intervention, but lack<br />

of a functional biochemical assay has limited efforts<br />

to discover compounds that target this essential viral<br />

protein. To date, only one compound targeting NS5A<br />

has been advanced to clinical trials. By applying a<br />

chemistry technology known as Diversity Oriented<br />

Synthesis and phenotypic screening against the<br />

cell-based HCV replicon, we have identified and<br />

optimized novel, selective and highly potent small<br />

molecule inhibitors of HCV replication that appear to<br />

act on NS5A and have favorable pharmacokinetic and<br />

pharmacodynamic properties.<br />

do not affect the growth of a range of other RNA<br />

and DNA viruses in culture. Transient treatment<br />

of replicon cells results in a rapid, > 6-log reduction<br />

in HCV RNA with no rebound following compound<br />

removal. Biochemical and genetic studies suggest<br />

that these molecules interact with domain I of NS5A.<br />

Compounds administered orally to rats at 25 mg/kg<br />

preferentially accumulate in liver, achieving C max<br />

of<br />

1-10 µg/g and C 8hr<br />

(a surrogate for C min<br />

) of 0.25 – 5 µg/g,<br />

hundreds of times above the replicon EC 90<br />

, and have<br />

elimination half-lives of 4-11 hr, suggesting that they<br />

may be suitable for b.i.d. dosing. No adverse clinical,<br />

hematologic, clinical chemistry or histopathologic<br />

changes were observed in rats receiving 250 mg/kg/d<br />

of leads in multiple-dose toxicology studies. Leads are<br />

not active in the Ames test or in assays for inhibition<br />

of major CYP-450 enzymes or hERG. Analysis of<br />

the metabolism of these compounds in microsomes<br />

and animals indicates that they are very stable, with<br />

metabolic half-lives in the range of hours, less than<br />

10% conversion to oxidative metabolites and less<br />

than 1% conversion to glucouronides.<br />

Conclusion: These data support the hypothesis<br />

that NS5A is a pharmacologically tractable target<br />

for blocking HCV replication and identify a family<br />

of novel small molecule compounds that target<br />

NS5A and, as such, could complement protease and<br />

polymerase inhibitors currently in development for<br />

treatment of HCV infection.<br />

Methods and Results: Lead molecules have<br />

nanomolar EC 50<br />

s against genotype 1a and 1b<br />

replicons, are active against replicons that are<br />

resistant to protease and polymerase inhibitors, are<br />

not toxic to a wide range of cultured cell lines and<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 111


Optimizing the Outcome of<br />

Therapeutics for HBV and HCV<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 113


Abstract 102<br />

Measuring and Mapping<br />

Partnership Integration for<br />

Optimizing Assessment and<br />

Treatment of Hepatitis C<br />

G Butt 1, 2 , W Hill 1 , L McGuinness 1 , and M Krajden 1, 2<br />

1 British Columbia Centre for Disease Control, Vancouver,<br />

British Columbia, Canada; 2 University of British<br />

Columbia, Vancouver, British Columbia, Canada<br />

Background: In response to the hepatitis C (HCV)<br />

epidemic in British Columbia, collaborative projects<br />

were established in four geographic areas with high<br />

prevalence rates: Campbell River, Kamloops, Prince<br />

George and Surrey. Service delivery began with<br />

initial partnerships between public health nurses<br />

and local physicians. Nurse leadership broadened<br />

the partnerships to include other disciplines. The<br />

resulting model of service delivery provided integrated<br />

prevention and care services linked through informal<br />

interprofessional partnerships. A recent evaluation<br />

of HCV treatment outcomes from the project sites<br />

demonstrated viral clearance rates comparable to<br />

published clinical trials. The aim of this study is to<br />

describe and measure the breadth, depth and extent<br />

of interprofessional partnerships for each project<br />

site.<br />

Methods: Leaders from each project site developed<br />

a list of their inter- and intra-agency partners with<br />

whom they had contact within six months. Partners<br />

were mailed a self-adminstered survey package that<br />

included the Integration of Human Services Measure ©<br />

which asked them to rate their present and desired<br />

level of service integration with each member of the<br />

partner list using a 5-point Likert scale. Descriptive<br />

statistics were calculated for each partner. The<br />

geographic location and mean scores for present and<br />

desired level of integration were displayed on a map<br />

of BC using GIS mapping techniques. Ethics approvals<br />

were obtained from revelant ethics review boards.<br />

Results: Survey response rates ranged from 34%<br />

to 53%. The number of partners ranged from 29 to<br />

68. Public health, provincial government agencies<br />

and specialist physicians had higher mean scores for<br />

present and future integration. Three sites operating<br />

for >5 years had the greatest number and diversity<br />

of partners and the least difference between current<br />

and desired integration mean ratings (Kamloops<br />

[difference=0.22], Prince George [difference=0.12]<br />

Campbell River [difference=0.37] versus Surrey<br />

[difference=0.45]). The majority of partners were<br />

located near the local project sites, however, all sites<br />

had linkages with other project sites, province-wide<br />

partnerships, and some national partnerships.<br />

Conclusion: This study demonstrates the diverse<br />

number and type of resources that are required to<br />

provide appropriate assessment and treatment for<br />

HCV. GIS mapping provided an effective mechanism<br />

to show the broad range of informally-linked<br />

partnerships. Therapeutic outcomes similar to<br />

research trials were achieved in partnerships with<br />

low current mean integration scores that were close<br />

to their desired state of integration.<br />

Abstract 103<br />

Development of a Novel Mouse<br />

Model to Evaluate Single and<br />

Combination Therapy Against<br />

Hepatitis B Virus<br />

MA Feitelson 1 , MM Clayton 1 , B Sun 1 , and RF Schinazi 2<br />

1 Department of Biology, College of Science and<br />

Technology, Temple University, Philadelphia, PA, USA;<br />

2 Emory University/VA Medical Center, Decatur, GA, USA<br />

BACKGROUND: Woodchuck hepatitis virus infected<br />

woodchucks have been used for preclinical<br />

development of drugs against hepatitis B virus<br />

(HBV). However, there is no simple in vivo model to<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 115


evaluate small amounts of compounds against wild<br />

type HBV. In addition, drug resistance is a problem<br />

in the development and application of new drugs<br />

against HBV infection.<br />

METHODS: To develop a model that addresses<br />

these limitations, HepAD38 cells, in which HBV<br />

replication is regulated by tetracycline, were grown as<br />

subcutaneous tumors in nude mice. Mice developing<br />

viremia were then left untreated, given tetracycline<br />

in the drinking water, and in some mice, tetracycline<br />

was replaced with PBS, lamivudine (3TC), clevudine<br />

(CLV), tenofovir dipivoxil fumarate (TDF), or CLV<br />

plus TDF combination therapy. Virus DNA titers<br />

were measured by real-time PCR during and after<br />

drug treatment. Toxicity was monitored by alanine<br />

amino transferase (ALT) measurements and by tumor<br />

weight in treated and control animals.<br />

RESULTS: In water fed and PBS injected mice, virus<br />

titers reached to ~10 9 copies/ml serum within 35 days<br />

of HepAD38 injection, while in tetracycline treated<br />

mice, virus titers remained at 10 4 -10 5 . HBV DNA levels<br />

were suppressed by 3TC, TDF and CLV, as expected,<br />

with the latter two drugs showing more sustained<br />

virus suppression compared to 3TC. Combination<br />

therapy with up to 27 fold lower concentrations of<br />

CLV plus TDF was much more effective than either<br />

drug alone in suppressing virus titer, which remained<br />

depressed for at least 3 weeks after the end of<br />

treatment. There was no demonstrable toxicity to<br />

HepAD38 cells in drug treated mice.<br />

CONCLUSIONS: Hence, a robust tetracycline controlled<br />

system for HBV replication in vivo has been<br />

demonstrated, validated with single drugs against<br />

HBV, and shown to be useful in assessing combination<br />

therapy. This system will be useful for preclinical<br />

assessment of small amounts of other single or<br />

multiple compounds against HBV in vivo before<br />

expensive human trials are initiated.<br />

Abstract 104<br />

Association of Pretreatment<br />

Serum Interferon-γ-inducible<br />

Protein 10 (Ip-10) Levels with<br />

Sustained Virological Response<br />

to Peginterferon Plus Ribavirin<br />

Therapy for Chronic Hepatitis<br />

C (CHC) with Virus Genotype 1<br />

Infection<br />

M Fukuda 1 , H Yatsuhashi 1 , R Nakao 1 , S Hashimoto 1 ,<br />

A Nishikawa 1 , N Hai 1 , M Tateyama 1 , Y Motoyoshi 1 ,<br />

S Nagaoka 1 , N Taura 1 , S Abiru 1 , K Yano 1 , A Komori 1 ,<br />

K Migita 1 , H Fujioka 1 , H Sakai 2 , E Takezaki 3 ,<br />

H Morimoto 4 , T Muro 5 , Y Hijioka 6 , M Yagura 7 ,<br />

N Masaki 8 , and H Ishibashi 1<br />

1 National NHO Nagasaki Medical Center, Clinical<br />

Research Center, Nagasaki, Japan; 2 National NHO<br />

Beppu Medical Center, Oita, Japan; 3 National NHO<br />

Higashihiroshima Medical Center, Hiroshima, Japan;<br />

4 National NHO Kanazawa Medical Center, Ishikawa,<br />

Japan; 5 National NHO Oita Medical Center, Oita, Japan;<br />

6 National NHO Osaka-Minami Medical Center, Osaka,<br />

Japan; 7 National NHO Tokyo Hospital, Tokyo, Japan;<br />

8 International Medical Center of Japan, Tokyo, Japan<br />

Background and aim: Interferon-γ inducible<br />

protein 10 kDa (IP-10 or CXCL10) is a CXC chemokine<br />

that, unlike other CXC chemokines, lacks chemotactic<br />

activity for neutrophils, but rather targets<br />

T lymphocytes, NK cells, and monocytes. Recently,<br />

baseline levels of IP-10 before the initiation of<br />

therapeutic intervention for hepatitis C virus (HCV)<br />

infection were reported to have close relationship to<br />

a sustained virological response (SVR) in Caucasian<br />

patients with HCV genotype 1 infection (Lagging M, et<br />

al, Hepatology 2006). Such molecules which associate<br />

with IFN response may have a different behavior<br />

among ethnic groups. We aimed this study to examine<br />

a possible association between the serum IP-10 level<br />

and virological response to the peg-interferon plus<br />

ribavirin (PegIFN/RBV) therapy in Japanese patients<br />

with CHC infected with genotype 1 HCV.<br />

116 Global Antiviral Journal Volume 3, Supplement 2


Methods and patients: Serum IP-10 levels were<br />

determined by enzyme linked immunosorbent assay.<br />

Serum IP-10 levels were measured in 40 healthy<br />

persons and 402 patients (202 men (50.2%) and 200<br />

women (49.8%); mean age 57.3 years old (SD; 10.2,<br />

range 17-79)) who initiated PegIFN/RBV therapy.<br />

The fibrosis stage and the grade of inflammation were<br />

determined in liver biopsy specimens taken before<br />

therapy.<br />

Results: IP-10 levels in 402 CHC patients were<br />

significantly higher than those in 40 healthy persons<br />

(522.8 (373.7) vs. 97.9 (24.9) pg/ml, respectively;<br />

P


mean attendance being 15 subjects per week (range<br />

3-32). Overall, 10 (8%) did not medically qualify for<br />

treatment, 39 (30%) were lost to follow-up and 8<br />

(6%) had completed or initiated treatment for HCV<br />

infection prior to attending the group. We observed a<br />

high uptake of HCV treatment among attendees, with<br />

30% of subjects (39/129) currently under evaluation<br />

and 26% (33/129) having initiated or completed<br />

treatment for HCV infection. In a comparison of<br />

subjects that had initiated or completed treatment<br />

for HCV infection (n=33) and those lost to follow<br />

up (n=39), those having received treatment for<br />

HCV infection had a higher median attendance [34<br />

meetings (Interquartile range, IQR = 11-33) vs. 2<br />

meetings (IQR=1-3, P3 clinic visits (97% vs. 23%, P


higher (P


Abstract 108<br />

Changes of IFN-inducible Gene<br />

(IP-10, PKR, MxA) Expressions<br />

During Pegylated Interferon Alfa-<br />

2b Plus Ribavirin Therapy in HCV<br />

Genotype 1 Infected Japanese<br />

Patients<br />

R Nakao, H Yatsuhashi, M Fukuda, A Nishikawa,<br />

S Hashimoto, N Hai, M Tateyama, Y Motoyoshi,<br />

S Nagaoka, N Taura, K Yanagi, K Yano, S Abiru,<br />

A Komori, H Fujioka, K Migita, M Nakamura,<br />

and H Ishibashi<br />

Clinical Research Center, National Hospital Organization<br />

Nagasaki Medical Center, Omura, Nagasaki, Japan<br />

BACKGROUND: Recently, it has been reported that<br />

pretreatment interferon(IFN)-gamma-inducible protein<br />

10 kDa (IP-10 or CXCL10) might be a predictive<br />

factor for sustained viral response to PEG-IFN plus<br />

RBV therapy in patients infected with hepatitis C virus<br />

(HCV) genotype 1. In order to clarify the role of IP-10<br />

in the treatment of IFN, we evaluated the changes of<br />

IFN-related gene expression during treatment and<br />

examined its relationship with the viral response to<br />

IFN.<br />

METHODS: A total of 81 HCV genotype 1 patients<br />

treated with PEG-IFN alfa-2b plus RBV. Serum IP-<br />

10 levels were measured by ELISA before therapy,<br />

during treatment (week 1, 2, 4, 12, and 24), end of<br />

treatment and 24 weeks after cessation of therapy. In<br />

addition, we examined IP-10, IFN receptor (IFNAR2),<br />

myxovirus resistance-A (MxA), RNA-dependent<br />

protein kinase (PKR), IFN regulatory factor-1 (IRF1),<br />

IRF2 (IRF2) and signal transducers and activators<br />

of transcription factor (STAT3) gene expression in<br />

peripheral blood mononuclear cells in 58 patients.<br />

The primary study end point was the early virologic<br />

response (EVR) – an undetectable serum HCV-RNA<br />

level at treatment week 12.<br />

patients with non-EVR were significantly higher than<br />

those with EVR (pretreatment: 486 v 384 pg/ml, P <<br />

0.05, at week 1: 645 v 428 pg/ml, P < 0.01, at weeks 2:<br />

444 v 318 pg/ml, P < 0.05, post-treatment: 418 v 186<br />

pg/ml, P < 0.001, respectively). The expression levels<br />

of IFNAR2, IRF1, IRF2 and STAT3 were significantly<br />

down-regulated by PEG-IFN plus RBV therapy<br />

while the expression levels of MxA and PKR were<br />

significantly up-regulated. At pretreatment, there<br />

was no difference in IFN-related gene expression<br />

between the EVR group and the non-EVR group. At<br />

week 2 compared with baseline, the change of PKR<br />

and IP-10 expression elevated significantly higher in<br />

non-EVR than in EVR (PKR: 2.5 v 1.7 fold, P < 0.05,<br />

IP-10: 1.0 v 0.7 fold, P < 0.05).<br />

CONCLUSIONS: Unexpectedly, the non-EVR group<br />

had a stronger response of IFN-related molecules<br />

upon PEG-IFN plus RBV administration than EVR<br />

group. It has been reported that elevated serum IP-<br />

10 levels were significantly associated with a poor<br />

response to antiviral therapy (J Infect Dis 2006,<br />

Hepatology 2006, Gut 2006). On the other hand, in<br />

long-term IFN-gamma treatment, suppression of IFNalfa<br />

signaling is mediated partly through elevation in<br />

STAT1 protein expression. These high levels of IFNgamma<br />

and STAT1 protein may suppress the effect<br />

of IFN-alfa therapy and, consequently contribute to<br />

IFN-alfa treatment failure in CHC patients (Biochem.<br />

J. 2004). We suggest that resistance to PEG-IFN plus<br />

RBV therapy may not be due to a disturbance in IFN<br />

cell signaling, but high levels of IP-10 and IFN-gamma<br />

negatively regulate IFN-alfa-activated signals.<br />

RESULTS: At pretreatment, week 1, 2 and 24 weeks<br />

after cessation of therapy, serum IP-10 levels in<br />

120 Global Antiviral Journal Volume 3, Supplement 2


Abstract 109<br />

Baseline Characteristics and Early<br />

Virologic Response to Telbivudine<br />

Predict 2‐Year Outcomes for<br />

Patients with HBeAg-negative<br />

Chronic Hepatitis B<br />

T Poynard 1 , GV Papatheodoridis 2 , M Tong 3 , N Tsai 4 ,<br />

and M Buti 5<br />

1 Groupe Hospitalier Pitié-Salpêtrière, Paris, France;<br />

2 Athens University Medical School, Athens, Greece;<br />

3 Huntington Medical Research Institutes, Pasadena, CA,<br />

USA; 4 John A Burns School of Medicine, Honolulu, HI,<br />

USA; 5 Hospital Valle de Hebrón, Barcelona, Spain<br />

BACKGROUND: The ability to predict long-term<br />

treatment response based on baseline demographics<br />

and early responses to therapy may help physicians<br />

individualize patient management and optimize<br />

outcomes in chronic hepatitis B. For HBeAg-negative<br />

patients, trends suggest better outcomes for patients<br />

with low baseline HBV DNA levels (


Abstract 110<br />

Predictors of Sustained Viral<br />

Response in the Retreatment of<br />

Previous Interferon/Ribavirin<br />

Nonresponders: Results from the<br />

EPIC 3 Program<br />

T Poynard 1 , E Schiff2 , R Terg 3 , R Moreno Otero 4 ,<br />

S Flamm 5 , W Schmidt 6 , T Berg 7 , F Goncales Jr 8 ,<br />

J Heathcote 9 , M Diago 10 , T McGarrity 11 , P Bedossa 12 ,<br />

W Deng 12 , P Mukhopadhyay 12 , L Griffel 12 ,<br />

M Burroughs 12 , C Brass 12 , and J K Albrecht 12<br />

1 Service d’hepatologie, Hôpital La Pitié Salpêtrière, Paris,<br />

France; 2 University of Miami School of Medicine, Miami,<br />

Florida, USA; 3 Hospital Municipal de Gastroenterologie<br />

Dr. Bonorino Udaondo, Capital Federal, Argentina;<br />

4 Hospital Universitario de la Princesa, Madrid, Spain;<br />

5 Northwestern University, Chicago, Illinois, USA;<br />

6 University of Iowa Hospitals and Clinics, Iowa City,<br />

Iowa, USA; 7 Virchow Klinikum der Charite, Berlin,<br />

Germany; 8 Hospital das Clinicas da Unicamp Cidade<br />

Universitaria, Campinas, Brazil; 9 University Health<br />

Network, Toronto, Ontario, Canada; 10 Hospital General<br />

Universitario de Valencia, Valencia, Spain; 11 Milton S.<br />

Hershey Medical Center, Hershey, Pennsylvania, USA;<br />

12 Schering-Plough Research Institute, Kenilworth, New<br />

Jersey, USA<br />

BACKGROUND: EPIC 3 includes a prospective<br />

trial designed to assess the efficacy and safety of<br />

retreatment of subjects with chronic hepatitis C and<br />

fibrosis (METAVIR F2-F4) who were nonresponsive<br />

to previous treatment with any interferon (IFN) alfa<br />

plus ribavirin with peginterferon (PEG-IFN) alfa-2b<br />

and ribavirin. The overall sustained virologic response<br />

(SVR) rate was 23% in these subjects and 57% among<br />

those with undetectable hepatitis C virus (HCV) RNA<br />

at treatment week (TW) 12. The aim of this analysis<br />

was to define predictors of SVR in these subjects.<br />

TW12, TW24, and TW48 and at follow-up weeks 12<br />

and 24 using a quantitative TaqMan assay (Schering-<br />

Plough Research Institute, Kenilworth, New Jersey,<br />

USA; lower limit of detection 125 IU/mL).<br />

RESULTS: Undetectable HCV RNA at TW12 was the<br />

most important predictor of SVR; 57% of subjects<br />

with undetectable HCV RNA at TW12 attained SVR.<br />

In contrast, only 6% of those with a 2 log 10<br />

or more<br />

drop in HCV RNA and with detectable virus at TW12<br />

attained SVR; no subjects with less than a 2 log 10<br />

drop at TW12 attained SVR. Results of a multivariate<br />

analysis indicate that other important predictors of<br />

SVR were fibrosis score (F2, 30% SVR; F3, 25% SVR;<br />

F4 17% SVR; P


Abstract 111<br />

Addition of Lamivudine to<br />

Patients with Chronic Hepatitis<br />

B who are Viremic on Adefovir<br />

Dipivoxil Lowers Plasma HBV<br />

DNA Levels<br />

M Tong 1 and J Ryan 2<br />

1 Huntington Medical Research Institute, Pasadena, CA,<br />

USA; 2 Gilead Sciences, Foster City, CA, USA<br />

Background: There is limited information on<br />

whether it is beneficial to add lamivudine (LAM)<br />

to patients with chronic hepatitis B (CHB) who are<br />

viremic while receiving adefovir dipivoxil (ADV)<br />

monotherapy.<br />

Methods: We performed a retrospective search of<br />

the HMRI (Huntington Medical Research Institute)<br />

database to identify viremic patients (HBV DNA ≥<br />

3 log 10<br />

copies/mL by PCR-based assay) who received ≥<br />

6 months of treatment with ADV, and who had LAM<br />

added to their treatment. Patients with or without<br />

LAM-resistance were included. The primary analysis<br />

was change in HBV DNA at 6 months after adding<br />

LAM; we also determined the proportion of patients<br />

with undetectable HBV DNA, and e-antigen loss or<br />

seroconversion at any time during the follow-up<br />

period.<br />

Results: 15 patients were identified who met<br />

inclusion criteria; 12 patients (80%) had a prior history<br />

of LAM use and 9 patients (60%) were LAM-resistant<br />

at the time of LAM addition (baseline). Median age<br />

was 50 yrs, 80% were male, 87% were Asian; 40% were<br />

HBeAg-positive. Median HBV DNA prior to starting<br />

ADV monotherapy was 6.4 log 10<br />

copies/mL. At the<br />

time LAM was added to ADV, median HBV DNA was<br />

4.7 log 10<br />

copies/mL, and patients had received ADV<br />

monotherapy for a median duration of 16 months.<br />

At 6 months following the addition of LAM, median<br />

HBV DNA declined by 2.1 log 10<br />

copies/mL; all<br />

patients became undetectable (LLD < 160 copies/mL)<br />

after a median of 6 months follow-up. HBeAg loss/<br />

seroconversion was seen in 40% of HBeAg-positive<br />

patients. Responses in patients with LAM-resistance<br />

were similar to those in patients without evidence of<br />

resistance.<br />

Conclusions: In this small cohort, adding LAM<br />

to ADV in patients with persistent viremia on ADV<br />

monotherapy appeared to be effective in controlling<br />

HBV replication, regardless of the presence of LAMresistance.<br />

Abstract 112<br />

A Systematic Review of the<br />

Effectiveness of Pegylated<br />

Interferon, Lamivudine, Adefovir<br />

and Entecavir for the Treatment<br />

of Hepatitis B<br />

G Woo 1 , Y Nishikawa, J Heathcote 1,2 , M Sherman 2 ,<br />

T Einarson 1 , W Ungar 1,3 , M Krahn 1,2,4<br />

1 University of Toronto, Ontario, Canada; 2 University<br />

Health Network, Ontario, Canada; 3 Hospital for Sick<br />

Children Research Institute, Ontario, Canada;<br />

4 Toronto Health Economics and Technology Assessment<br />

Collaborative, Ontario, Canada<br />

Background: Treatment options for chronic<br />

hepatitis B (CHB) are constantly evolving. An upto-date<br />

and comprehensive analysis comparing<br />

the effectiveness of all available treatments is not<br />

currently available.<br />

Aims: To systematically review the effectiveness<br />

of pegylated interferon (PEG), lamivudine (LAM),<br />

adefovir (ADF) and entecavir (ENT) in treating CHB.<br />

Methods: Pubmed, Embase, Cochrane, and Econlit<br />

were searched for randomized controlled trials<br />

assessing the efficacy of the selected drugs for treating<br />

CHB published in the English language from the dates<br />

of inception to the end of December 2006. Patients<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 123


were considered to have CHB if they had elevated<br />

ALT levels and active viral replication. Monotherapy,<br />

combination and sequential therapies were included.<br />

The treatment duration was one year. The search<br />

was performed with the aid of a librarian. Two<br />

independent reviewers independently assessed the<br />

relevance of the studies; discrepancies were solved<br />

through debate and assessment by a third reviewer.<br />

We included studies that documented: patient<br />

randomization, patient baseline characteristics,<br />

defined eligibility criteria, patient blinding, adequate<br />

information to assess one of our targeted clinical<br />

endpoints and reporting of drop-outs within the<br />

one year treatment period. Among trials that met<br />

our inclusion criteria, we abstracted data describing<br />

normalization of ALT, HBV DNA, sustained<br />

biochemical response, HBeAg seroconversion,<br />

histological improvement, drop-outs and adverse<br />

events. Data was collected and checked by two<br />

independent reviewers. Intention-to-treat data<br />

were combined using a random-effects metaanalysis,<br />

with missing data considered as treatment<br />

failures. Outcomes were expressed as relative risks<br />

versus placebo or active drug with 95% confidence<br />

intervals.<br />

Results: The initial search yielded 2064 references,<br />

127 were excluded due to inadequate blinding,<br />

allocation concealment, randomization and reporting<br />

of outcomes; 20 studies were included. Trials involved<br />

5573 patients (4121 males, 1309 females), ranging<br />

in size from 200-814 patients. Mean age was 40.7.<br />

Monotherapy comparisons available were LAM,<br />

ADF, and ENT versus placebo, and LAM versus PEG,<br />

ADF and ENT. Eleven trials studied HBeAg-positive<br />

patients, four trials studied HBeAg-negative patients,<br />

and four trials studied both. Due to small numbers of<br />

trials for comparison led to pooling of HBeAg-positive<br />

and HBeAg-negative studies.<br />

No treatment was superior for all outcome measures.<br />

Monotherapy was found to be superior to placebo.<br />

Comparisons of single drugs favored treatment with<br />

ADF or ENT over LAM or PEG. In direct comparisons,<br />

LAM was superior to PEG with better clinical<br />

outcomes and fewer adverse events and patient<br />

dropouts. Combination and sequential treatments<br />

were not superior, but comparisons were limited by<br />

our one-year follow-up.<br />

Conclusion: Monotherapy with ADF or ENT are<br />

the most attractive treatment options within the first<br />

year of treatment. Further research on combination<br />

and sequential therapies may provide better options<br />

but presently insufficient evidence exists to support<br />

this approach.<br />

Abstract 113<br />

Safety Recommendations for<br />

Laboratory Values in Specific<br />

Product Characteristics (SPC) of<br />

Peginterferon Alfa-2a (PEG) and<br />

Ribavirin (RBV) - What Happens<br />

Under Real Life Conditions?<br />

E Zehnter 1 , S Mauss 2 , K Boeker 3 , T Lutz 4 , S Racky 5 ,<br />

W Schmidt 6 , R Ullrich 7 , R Heyne 8 , A Schober 9 , C John 10 ,<br />

KH Hey 11 , B Möller 8 , B Bokemeyer 12 , B Kallinowski 13 ,<br />

S Pape 14 , U Alshuth 15 , and D Hüppe 16<br />

1 Center of Gastroenterology, Dortmund, Germany;<br />

2 Center of HIV and Hepatogastroententerology,<br />

Duesseldorf, Germany; 3 Center of Gastroenterology,<br />

Hannover, Germany; 4 Center of Infectiology, Frankfurt,<br />

Germany; 5 Center of Gastroenterology, Bad Schwalbach,<br />

Germany; 6 Center of Gastroenterology, Berlin, Germany;<br />

7 Center of Gastroenterology, Krefeld, Germany;<br />

8 Livercenter, Berlin, Germany; 9 Center of<br />

Gastroenterology, Goettingen, Germany; 10 Center of<br />

Gastroenterology, Berlin, Germany; 11 General Practice,<br />

Paderborn, Germany; 12 Center of Gastroenterology,<br />

Minden, Germany; 13 Center of Gastroenterology,<br />

Schwetzingen, Germany; 14 Center of Gastroenterology,<br />

Paderborn, Germany; 15 BU Hepatitis/HIV/Infectiology,<br />

Roche Pharma AG, Grenzach-Wyhlen, Germany;<br />

16 Center of Gastroenterology, Herne, Germany<br />

BACKGROUND: Recommendations of SPCs derived<br />

from forced conditions in pivotal trials. A very<br />

important part concerns haematological laboratory<br />

data at start and during therapy. Information about<br />

adherence to SPC recommendations are missing.<br />

124 Global Antiviral Journal Volume 3, Supplement 2


METHODS: Until May 2006 data of 4377 patients<br />

in different phases of CHC treatment with PEG<br />

and RBV were recorded in a German observational<br />

trial conducted by BNG and Roche. In the following<br />

treatment behaviour is described, if laboratory<br />

data achieve recommended cut-off values of SPC.<br />

RESULTS: A small part of pts had baseline values<br />

under recommended numbers of haematological<br />

parameters: Neutrophils (Neu)


81/2234 (3.6%) by histology (H). In 310 patients<br />

(3.0%) cirrhosis was clinically diagnosed (C) and<br />

classified according to Child Pugh: A 86.5%, B 10.0%,<br />

C 3.5%. Patients diagnosed by sonography were on<br />

average 58.2 yrs., 60.6% male, with BMI 26.3 kg/<br />

m², thrombocytes 137,989 /µl, duration of infection<br />

19.2 yrs., source of infection (multiple answers<br />

possible): blood products 31.4%, IVDU 23.7%,<br />

medical intervention 8.7%, unknown 35.3%. Current<br />

alcohol abuse was reported for 12.2% of patients.<br />

Distribution of genotypes: G1 75.3%, G2/3 21.1%, G4<br />

3.1%. Treatment-rate with Peginterferon alfa-2a and<br />

Ribavirin was (according to diagnosis tool): 31.4%<br />

(S), 75.5% (H), 42.6% (C). SVR rate of patients with<br />

cirrhosis (S) was 37.3% (28/75). Reasons against a<br />

therapy were (S): decompensated disease 33.6%, other<br />

therapy 10.2%, concomitant disease/continuous drug<br />

abuse 25.7%, patient’s request 22.2%, age 8.3%.<br />

CONCLUSIONS: About 3-5% of the described patient<br />

group already had a liver cirrhosis at screening. The<br />

patients are older and have been infected for longer<br />

than average. Most common way of infection was<br />

medical intervention. Although these patients are<br />

in urgent need of hepatitis C therapy, more than<br />

1/5 cannot be convinced to start. Success rate of<br />

therapy in this real-life setting was similar to the<br />

numbers observed in clinical studies. However, due<br />

to decompensation or other diseases, for the majority<br />

of patients interferon therapy is not an option any<br />

more.<br />

126 Global Antiviral Journal Volume 3, Supplement 2


Late Breaker<br />

Session<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 127


Abstract 115<br />

Long Term Follow-Up of HCV<br />

Patients Completing Peg-IFN Plus<br />

Ribavirin Treatment – Is There a<br />

Cure?<br />

MP Manns<br />

Medical School of Hannover, Hannover, Germany<br />

The goal of treatment in chronic hepatitis C is to<br />

achieve undetectable HCV RNA in serum 24 weeks<br />

after the end of therapy. It has been a debate whether<br />

this means cure of this chronic viral infection.<br />

Information on HCV RNA longterm after the end<br />

of therapy is limited. Therefore the information is<br />

valuable for long term follow up studies of patients<br />

that were treated as part of large pivotal trials<br />

with interferon based therapies. Data are available<br />

from several pivotal trials with pegylated and nonpegylated<br />

interferon alpha 2a and 2b alone or in<br />

combination with ribavirin.<br />

Non-pegylated interferon alpha 2b (McHutchison<br />

et al, EASL 2006) : 1071 treatment naïve patients<br />

and patients relapsed after the end of therapy were<br />

treated with interferon alpha 2b (Intron A) +/-<br />

ribavirin (Rebetol) and who completed 24 weeks of<br />

follow up were recruited. HCV RNA was measured by<br />

PCR (NGI PCR with sensitivity of 100 copies/ml and<br />

after 03/2001 with Taqman SP assay and a sensitivity<br />

of 29 IU/ml). Of the 1071 pts enrolled in the long<br />

term study 492 had achieved SVR and 579 were<br />

non-responders (NR). 61 % of SVR and 28 % of NR<br />

patients completed 5 years of follow up with a median<br />

duration of 283 and 131 weeks, respectively. Only 5<br />

of the 492 SVR patients had a definite relapse on long<br />

term follow up. There were an additional 7 patients<br />

with possible recurrent disease with inconsistent<br />

HCV RNA results. Thus the probability of SVR for the<br />

5 year posttreatment follow up period was between<br />

97 – 99 % for these patients.<br />

Pegylated Interferon alpha 2b (Schering-Plough,<br />

personal communication; Manns et al, submitted):<br />

567/1695 (33 %) of patients from two pivotal trials<br />

(Lindsay et al , Hepatology, 2001; Manns et al, Lancet<br />

2001) treated with Peg-IFN alpha 2b +/- ribavirin<br />

who completed 24 week follow up were assessed<br />

annually for up to 5 years for evidence of disease<br />

progression and sustained HCV RNA negativity<br />

(Taqman SP PCR, sensitivity 29 IU/ml ). Three<br />

SR subjects relapsed during the 5 year FU period,<br />

all within the first two years. The Kaplan-Meier<br />

estimate for continued SVR over 5 years is 99%.<br />

Pegylated Interferon alpha 2a (Swain et al , EASL 2007):<br />

997 patients treated with pegylated interferon alpha<br />

2a (Pegasys) alone or in combination with ribavirin<br />

(Rebetol) were followed for a mean of 4.1 years after<br />

the end of treatment. They were from 4 studies with<br />

monotherapy of PEG-IFNalpha2a with elevated ALT,<br />

4 studies with combination therapy and elevated<br />

ALT, 1 study with combination therapy and normal<br />

ALT, and 1 study with HCV/HIV co-infection treated<br />

with either mono- or combination therapy. 989/997<br />

patients (99%) stayed HCV RNA negative (range 0.4 –<br />

7 years) after treatment cessation. 8 patients became<br />

positive between 1.1 and 2.9 years after completing<br />

therapy. Six of these 8 patients had high viral load<br />

pre-treatment.<br />

Conclusions: For interferon alpha 2a and 2b,<br />

pegylated or non-pegylated, with or without ribavirin,<br />

HCV RNA negativity 24 weeks after the end of therapy<br />

is an excellent predictor of long term clearance<br />

of HCV RNA from serum. These data confirm the<br />

concept of cure in the treatment of chronic hepatitis<br />

C by interferon based therapies.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 129


Abstract 116<br />

Delta Hepatitis: Treatment<br />

Options for a Largely Neglected<br />

Disease<br />

MP Manns and H Wedemeyer<br />

Dept. of Gastroenterology, Hepatology and<br />

Endocrinology, Hannover Medical School,<br />

Carl-Neuberg-Straße 1, 30625 Hannover, Germany<br />

The hepatitis delta virus (HDV) is a satellite virus<br />

that requires co-infection with hepatitis B virus<br />

(HBV) for its replication. Delta hepatitis represents<br />

possibly the most severe form of chronic hepatitis.<br />

In chronically infected HBV carriers, HDV can result<br />

in fulminant acute hepatitis or severe chronic active<br />

hepatitis, progressing to cirrhosis in at least two<br />

thirds of patients at a younger age, and increasing the<br />

risk of hepatocellular cancer threefold and mortality<br />

twofold compared with chronic HBV infection alone.<br />

Worldwide, around 15 million people are infected<br />

with HDV and are therefore at risk of the potentially<br />

severe sequalae of infection. The epidemiology of<br />

HDV infection has changed in recent decades, with<br />

reports of decreased prevalence in some countries,<br />

linked to the introduction of vaccination against<br />

HBV. In contrast, the epidemiology of delta hepatitis<br />

had changed significantly in Germany with large<br />

numbers of patients migrating to Germany from<br />

Eastern Europe and countries of the former Soviet<br />

Union. Subsequently, the anti-HDV prevalence<br />

among HBsAg-positive individuals remained constant<br />

between 8% and 14% since 1997 at our center.<br />

Treatment options for delta hepatitis are limited.<br />

So far, no nucleoside or nucleotide analogue used<br />

for the treatment of viral hepatitis has shown any<br />

efficacy against HDV. Interferon alpha may cure delta<br />

hepatitis in single patients but requires prolonged<br />

administration with high doses. Three pilot studies<br />

with PEG-IFN alpha-2b (1.5 μg/kg/week) have<br />

demonstrated a sustained HDV-RNA response of<br />

17%-43%. We did investigate the efficacy of pegylatedinterferon<br />

alfa-2a and/or adefovir dipivoxil in 90<br />

patients with delta hepatitis recruited in Germany,<br />

Turkey and Greece treated for 48 weeks. An at least<br />

2xlog 10<br />

-decline of HDV-RNA was observed in 39%<br />

of patients treated with PEG-IFNa-2a plus adefovir,<br />

44% of patients treated with PEG-IFNa-2a and<br />

placebo and 8% treated with adefovir. A sustained<br />

HDV-RNA response was observerd in roughly one<br />

quarter of patients receiving PEG-IFN either alone<br />

or in combination with adefovir. Interestingly, the<br />

combination therapy group was superior in HBsAg<br />

reduction which may also have significant impacts<br />

for the treatment of HBV infection without delta<br />

hepatitis.<br />

In summary, delta hepatitis is a largely underestimated<br />

severe liver disease in Europe affecting mainly<br />

immigrants. PEG-IFN alpha treatment should be<br />

considered and the role of combination and prolonged<br />

therapies requires further investigation.<br />

Abstract 117<br />

Biochemical Mechanism of<br />

Entecavir as an Antiviral<br />

Polymerase Inhibitor<br />

RA Domaoal 1 , M McMahon 2 , CL Thio 2 , CM Bailey 1 ,<br />

J Tirado-Rives 4 , A Obikhod 3 , M Detorio 3 , KL Rapp 3 ,<br />

RF Siliciano 2 , RF Schinazi 3 , and KS Anderson 1<br />

1 The Yale University School of Medicine, Department<br />

of Pharmacology, New Haven, Connecticut 06520-<br />

8066, USA; 2 Department of Medicine, Johns Hopkins<br />

University School of Medicine, and Howard Hughes<br />

Medical Institute, Baltimore MD 21205, USA; 3 Emory<br />

University School of Medicine/Veterans Affairs Medical<br />

Center, Decatur, Georgia, 30033, USA; 4 Yale University,<br />

Department of Chemistry, New Haven, Connecticut<br />

06510, USA<br />

The novel 2’-deoxyguanosine analog Entecavir (ETV)<br />

is a potent inhibitor of hepatitis B virus (HBV)<br />

replication and is recommended for treatment in<br />

human immunodeficiency virus type 1 (HIV-1) and<br />

HBV co-infected patients because it had been reported<br />

that ETV is HBV-specific. Recent clinical observations,<br />

130 Global Antiviral Journal Volume 3, Supplement 2


however, have suggested that ETV may indeed<br />

demonstrate anti-HIV-1 activity. To investigate this<br />

question at a molecular level, kinetic studies were used<br />

to examine the interaction of ETVTP with wild type<br />

(WT) HIV-1 reverse transcriptase (RT) and the NRTI<br />

resistant mutation M184V. Using single turnover<br />

kinetic assays, we found that HIV-1 WT RT and<br />

M184V RT could use the activated ETV triphosphate<br />

metabolite as a substrate for incorporation. The<br />

mutant displayed a slower incorporation rate, a lower<br />

binding affinity and a lower incorporation efficiency<br />

with ETVTP compared to WT RT, suggesting a kinetic<br />

basis for resistance. Our results are supported by<br />

cell-based assays in primary human lymphocytes<br />

that show inhibition of WT HIV-1 replication by ETV<br />

and decreased susceptibility of the HIV-1 containing<br />

the M184V mutation. This study has important<br />

therapeutic implications as it establishes ETV as an<br />

inhibitor for HIV-1 RT and illustrates the mechanism<br />

of resistance by the M184V mutant.<br />

Abstract 118<br />

Advancing RNAi-based<br />

Therapeutic from Discovery to<br />

Clinic<br />

C Pachuk 1 and R Gish 2<br />

1 Nucleonics Inc., Horsham PA, USA; 2 California Pacific<br />

Medical Research Institute, San Francisco, CA, USA<br />

The presentation will provide a brief overview of<br />

Nucleonics’ RNA interference technology, preclinical<br />

and clinical development of its eiRNA-based anti-<br />

HBV therapeutic, NUCB1000. Unlike current small<br />

molecule HBV therapies, RNAi-based drug product<br />

NUCB1000 has been shown to suppress viral antigen<br />

expression in addition to inhibiting viral replication.<br />

NUCB1000 has been designed to be effective against<br />

all HBV genotypes and has demonstrated activity<br />

against known drug resistant mutants. Because<br />

NUCB1000 targets multiple RNA sequences, it is not<br />

predicted to select for viable escape mutants over the<br />

course of therapy. The presentation will detail product<br />

development, including delivery, efficacy, safety<br />

in preclinical models and the clinical development<br />

program. NUCB1000 is currently undergoing safety<br />

evaluations in Phase-1b clinical studies in CHB<br />

patients.<br />

Abstract 119<br />

Ultra-deep Pyrosequencing of<br />

HBV Quasispecies in Nucleoside<br />

Analog Treated and Untreated<br />

Patients<br />

S Margeridon-Thermet 1 , N Shulman 1 , A Ahmed 1 ,<br />

T Liu 1 , C Wang 1 , B Simen 2 , J Simons 2 , M Egholm 2 ,<br />

B Gharizadeh 3 , and RW Shafer 1<br />

1 Department of Medicine, Stanford University, Stanford,<br />

CA, USA; 2 454 Life Sciences, Branford, Connecticut, USA;<br />

3 Stanford Genome Technology Center, Stanford, CA,<br />

USA<br />

BACKGROUND: HBV nucleoside analog (NA)<br />

resistance has become increasingly common. There<br />

is a high level of cross-resistance among available<br />

NA with mutations selected by one NA contributing<br />

resistance to others. Because HBV is a quasispecies,<br />

we sought to determine whether it is possible to<br />

detect minor NA-resistant variants that are not<br />

detected by standard direct-PCR dideoxynucleoside<br />

(Sanger) sequencing.<br />

METHODS: 15 cryopreserved plasma samples from<br />

HBV-infected patients with well-characterized<br />

treatment histories were sequenced by Sanger<br />

sequencing and by ultra-deep pyrosequencing (UDPS;<br />

454 Life Sciences) using the FLX platform. For UDPS,<br />

four pairs of primers tailed with 454 adaptors and<br />

a patient-specific bar code were used to amplify<br />

(Expand High Fidelity PCR System) 1,226 bp of HBV<br />

encompassing the known NA-resistance mutations.<br />

A plasmid HBV clone was sequenced as a control to<br />

determine the sequencing error rate.<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 131


RESULTS: Plasma samples were obtained from<br />

4 treated and 11 untreated patients. The treated<br />

patients had received either 3TC (n=1), 3TC/adefovir<br />

(n=1), or 3TC/adefovir/entecavir (n=2). The median<br />

plasma HBV DNA was 6.7 log copies/ml (range: 5.2 to<br />

8.5 log copies/ml). A minimum of 400 viral templates<br />

was submitted for UDPS. Each of the 64 amplicons<br />

from the 16 HBV samples were sequenced on a single<br />

PicoTiter plate. A median 16,000 reads of >200<br />

nucleotides were obtained per HBV sample providing<br />

a median sequence coverage of 3,200 per bp (range:<br />

1,170 to 6,459). The single-read mismatch error rate<br />

was estimated to be 0.1% in non-homopolymeric<br />

regions and 0.45% in homopolymeric regions<br />

providing sensitivity for detecting minor variants of<br />

between 0.2% to 2.0% depending on the region and<br />

extent of sequence coverage.<br />

Three of four treated patients had drug-resistant<br />

variants detected by UDPS but not Sanger sequencing.<br />

In the 3TC-treated patient, L180M and M204V were<br />

detected by Sanger sequencing and UDPS; L173V<br />

was detected only by UDPS (16% of reads). In the<br />

3TC/adefovir-treated patients, N236T was detected<br />

only by UDPS (6% of reads). In one of the threedrug<br />

treated persons M204V/I and L180M/V were<br />

detected by UDPS and Sanger sequencing, but L80V<br />

(1.5% of reads) and A181T (0.8% of reads) were<br />

detected only by UDPS. Among the 11 untreated<br />

patients, low-levels (2% to 24%) of the accessory<br />

drug resistance mutations V214A and I233V were<br />

detected in 3 patients. G to A hypermutation, which<br />

has the potential to complicate sequence analysis, was<br />

present at low levels in several patients and validated<br />

by clonal sequencing.<br />

Conclusions: UDPS quantified known HBVresistance<br />

mutations at a level of sensitivity not<br />

previously possible. UDPS will provide new insight<br />

into HBV population genetics in NA-treated and<br />

untreated HBV-infected patients.<br />

132 Global Antiviral Journal Volume 3, Supplement 2


Author Index<br />

Author Abstract Page<br />

Afdhal, NH. ..................40.. . . . . . . . . . . . . . . . 41<br />

Alter, H. ...................... 3.. . . . . . . . . . . . . . . . . . 7<br />

Anderson, KS................117.. . . . . . . . . . . . . . . 130<br />

Ayres, A .. . . . . . . . . . . . . . . . . 79, 80.. . . . . . . . . . . . . 85, 86<br />

Balfe, P.......................54.. . . . . . . . . . . . . . . . 59<br />

Bartenschlager, R.. . . . . . . . . . . . . 04.. . . . . . . . . . . . . . . . . . 8<br />

Bassit, L.. . . . . . . . . . . . . . . . . . . . . 81.. . . . . . . . . . . . . . . . 87<br />

Blatt, LM.. . . . . . . . . . . . . . . . . . . . 58.. . . . . . . . . . . . . . . . 63<br />

Brass, V .. . . . . . . . . . . . . . . . . . . . . 07.. . . . . . . . . . . . . . . . 10<br />

Butt, G.. . . . . . . . . . . . . . . . . . . . . 102.. . . . . . . . . . . . . . . 115<br />

Chisari, FV....................19.. . . . . . . . . . . . . . . . 21<br />

Chu, CK .. . . . . . . . . . . . . . . . . . . . . 82.. . . . . . . . . . . . . . . . 88<br />

Chung, KW .. . . . . . . . . . . . . . . . . . 29.. . . . . . . . . . . . . . . . 30<br />

Crabbé, R.. . . . . . . . . . . . . . . . . . . . 56.. . . . . . . . . . . . . . . . 61<br />

Dash, S.. . . . . . . . . . . . . . . . . . 13, 14.. . . . . . . . . . . . . 14, 15<br />

Dayaram, YK.. . . . . . . . . . . . . . . . . 91.. . . . . . . . . . . . . . . . 99<br />

De Francesco, R...............24.. . . . . . . . . . . . . . . . 25<br />

De La Rosa, A .. . . . . . . . . . . . . . . . 83.. . . . . . . . . . . . . . . . 89<br />

Dennin, RH.. . . . . . . . . . . . . . . . . . 64.. . . . . . . . . . . . . . . . 68<br />

Di Bisceglie, AM.. . . . . . . . . . . . . . 39.. . . . . . . . . . . . . . . . 41<br />

Eroglu, C .. . . . . . . . . . . . . . . . . . . . 84.. . . . . . . . . . . . . . . . 90<br />

Evans, MJ .. . . . . . . . . . . . . . . . . . . 53.. . . . . . . . . . . . . . . . 59<br />

Ewart, G......................65.. . . . . . . . . . . . . . . . 69<br />

Feitelson, MA .. . . . . . . . . . . 66, 103.. . . . . . . . . . . . 70, 115<br />

Fenton, K.....................01.. . . . . . . . . . . . . . . . . . 3<br />

Flexner, CW...................98.. . . . . . . . . . . . . . . 107<br />

Fried, MW.. . . . . . . . . . . . . . . . . . . 77.. . . . . . . . . . . . . . . . 83<br />

Fukuda, M.. . . . . . . . . . . . . . . . . . 104.. . . . . . . . . . . . . . . 116<br />

Gastaminza, P.................15.. . . . . . . . . . . . . . . . 16<br />

Gish, R.. . . . . . . . . . . . . . . . . . . . . 118.. . . . . . . . . . . . . . . 131<br />

Glebe, D.. . . . . . . . . . . . . . . . . . . . . 47.. . . . . . . . . . . . . . . . 51<br />

Grakoui, A.. . . . . . . . . . . . . . . . . . . 06.. . . . . . . . . . . . . . . . . . 9<br />

Grebely, J....................105.. . . . . . . . . . . . . . . 117<br />

Hausman, DF .. . . . . . . . . . . . . . . . 67.. . . . . . . . . . . . . . . . 71<br />

Hostetler, KY.................26.. . . . . . . . . . . . . . . . 27<br />

Husa, P......................106.. . . . . . . . . . . . . . . 118<br />

Husova, L. ...................68.. . . . . . . . . . . . . . . . 72<br />

Jacobson, I. ..................23.. . . . . . . . . . . . . . . . 25<br />

Kim, HJ.. . . . . . . . . . . . . . . . . . . . 107.. . . . . . . . . . . . . . . 119<br />

Author Abstract Page<br />

Kiss, A. . . . . . . . . . . . . . . . . . . . . . . 57.. . . . . . . . . . . . . . . . 62<br />

Klade, CS.. . . . . . . . . . . . . . . . . . . . 48.. . . . . . . . . . . . . . . . 52<br />

Koike, K.. . . . . . . . . . . . . . . . . . . . . 49.. . . . . . . . . . . . . . . . 53<br />

Korba, BE.....................21.. . . . . . . . . . . . . . . . 23<br />

Koziel, MJ.. . . . . . . . . . . . . . . . . . . 44.. . . . . . . . . . . . . . . . 49<br />

Lau, GKK.. . . . . . . . . . . . . . . . 30, 43.. . . . . . . . . . . . . 30, 45<br />

Law, M.. . . . . . . . . . . . . . . . . . . . . . 45.. . . . . . . . . . . . . . . . 49<br />

Lee, CH......................31.. . . . . . . . . . . . . . . . 31<br />

Lefkowitz, EJ .. . . . . . . . . . . . . . . . 59.. . . . . . . . . . . . . . . . 64<br />

Lemon, SM .. . . . . . . . . . . . . . . . . . 18.. . . . . . . . . . . . . . . . 21<br />

Liang, TJ .. . . . . . . . . . . . . . . . . . . . 05.. . . . . . . . . . . . . . . . . . 8<br />

Lin, K .. . . . . . . . . . . . . . . . . . . . . . . 28.. . . . . . . . . . . . . . . . 29<br />

Lindsay, KL .. . . . . . . . . . . . . . . . . . 99.. . . . . . . . . . . . . . . 109<br />

Liu-Young, G.. . . . . . . . . . . . . . . . . 10.. . . . . . . . . . . . . . . . 11<br />

Locarnini, S.. . . . . . . . . . . . . . 02, 85.. . . . . . . . . . . . . . 3, 90<br />

Love, R.. . . . . . . . . . . . . . . . . . . . . . 69.. . . . . . . . . . . . . . . . 73<br />

Luscombe, CA.. . . . . . . . . . . . . . . . 32.. . . . . . . . . . . . . . . . 32<br />

Manns, MP .. . . . . . . . . . . . 115, 116.. . . . . . . . . . . 129, 130<br />

Marcellin, P.. . . . . . . . . . . . . . . . . . 63.. . . . . . . . . . . . . . . . 67<br />

Margeridon-Thermet, S........119.. . . . . . . . . . . . . . . 131<br />

Martinot-Peignoux, M..........11.. . . . . . . . . . . . . . . . 12<br />

Matsuura, Y...................27.. . . . . . . . . . . . . . . . 28<br />

McHutchison, JG.. . . . . . . . . 61, 70.. . . . . . . . . . . . . 65, 73<br />

McPhee, F .. . . . . . . . . . . . . . . . . . 100.. . . . . . . . . . . . . . . 110<br />

Morrey, JD...................71.. . . . . . . . . . . . . . . . 74<br />

Murakami, E.. . . . . . . . . . . . . . . . . 33.. . . . . . . . . . . . . . . . 32<br />

Nakao, R....................108.. . . . . . . . . . . . . . . 120<br />

Nelson, D.....................62.. . . . . . . . . . . . . . . . 66<br />

Neumann, AU.. . . . . . . . . . . . . . . . 22.. . . . . . . . . . . . . . . . 24<br />

Oh, J-W.. . . . . . . . . . . . . . . . . 72, 73.. . . . . . . . . . . . . 75, 76<br />

Olsen, DB....................60.. . . . . . . . . . . . . . . . 65<br />

Omata, M....................42.. . . . . . . . . . . . . . . . 44<br />

Park, SJ .. . . . . . . . . . . . . . . . . . . . . 16.. . . . . . . . . . . . . . . . 16<br />

Pachuk, C....................118.. . . . . . . . . . . . . . . 131<br />

Pedersen, IM.. . . . . . . . . . . . . . . . . 09.. . . . . . . . . . . . . . . . 11<br />

Perelson, AS..................08.. . . . . . . . . . . . . . . . 10<br />

Peters, MG....................88.. . . . . . . . . . . . . . . . 97<br />

Polyak, S.....................74.. . . . . . . . . . . . . . . . 77<br />

Poynard, T.. . . . . . . . . . 41, 109, 110.. . . . . . . . 43, 121, 122<br />

HEP DART 2007 - Frontiers in Drug Development for Viral Hepatitis 133


Author Index<br />

Author Abstract Page<br />

Ray, AS.......................97.. . . . . . . . . . . . . . . 107<br />

Reddy, KR .. . . . . . . . . . . . . . . . . . . 12.. . . . . . . . . . . . . . . . 13<br />

Ryan, J.. . . . . . . . . . . . . . . . . . . . . 111.. . . . . . . . . . . . . . . 123<br />

Sandrasagra, A .. . . . . . . . . . . . . . . 34.. . . . . . . . . . . . . . . . 33<br />

Shafer, RW. ..................20.. . . . . . . . . . . . . . . . 22<br />

Shimotohno, K.. . . . . . . . . . . . . . . 55.. . . . . . . . . . . . . . . . 60<br />

Shin, JW. ....................50.. . . . . . . . . . . . . . . . 53<br />

Simister, PC...................78.. . . . . . . . . . . . . . . . 84<br />

Smith, CA. ...................92.. . . . . . . . . . . . . . . 100<br />

Sozzi, T. ................. 86, 87.. . . . . . . . . . . . . 91, 92<br />

Swan, T. .....................90.. . . . . . . . . . . . . . . . 98<br />

Taylor, J.. . . . . . . . . . . . . . . . . . . . . 75.. . . . . . . . . . . . . . . . 77<br />

Temesgen, Z .. . . . . . . . . . . . . . . . . 89.. . . . . . . . . . . . . . . . 97<br />

Author Abstract Page<br />

Thommes, PA.................35.. . . . . . . . . . . . . . . . 34<br />

Tomaka, F .. . . . . . . . . 93, 94, 95, 96.. . 100, 101, 102, 103<br />

Tran, CV.................. 36, 37.. . . . . . . . . . . . . 35, 36<br />

Ujino, S......................38.. . . . . . . . . . . . . . . . 37<br />

Vaillant, A .. . . . . . . . . . . . . . . . . . . 25.. . . . . . . . . . . . . . . . 26<br />

Visvanathan, K.. . . . . . . . . . . . . . . 46.. . . . . . . . . . . . . . . . 50<br />

Wen, YM .. . . . . . . . . . . . . . . . . . . . 51.. . . . . . . . . . . . . . . . 54<br />

Wobbe, CR...................101.. . . . . . . . . . . . . . . 111<br />

Woo, G.. . . . . . . . . . . . . . . . . . . . . 112.. . . . . . . . . . . . . . . 123<br />

Yi, M.........................17.. . . . . . . . . . . . . . . . 17<br />

Zehnter, E .. . . . . . . . . . . . . 113, 114.. . . . . . . . . . . 124, 125<br />

Zheng, B.....................52.. . . . . . . . . . . . . . . . 55<br />

Zimmerman, KA...............76.. . . . . . . . . . . . . . . . 78<br />

134 Global Antiviral Journal Volume 3, Supplement 2


Informed Horizons presents...<br />

Upcoming Meetings<br />

HIV DRUG RESISTANCE<br />

w o r k s h o p<br />

basic principles & clinical implications<br />

June 10-14, 2008 • Sitges, Spain<br />

HIV<br />

<br />

2008<br />

XVII International HIV Drug<br />

Resistance Workshop:<br />

Basic Principles & Clinical Applications<br />

June 10-14, 2008<br />

Sitges, Spain<br />

This workshop is renowned for the quality of the<br />

data presented and the depth of the scientific<br />

interaction and debate.<br />

HIV DART 2008<br />

Frontiers in Drug Development for<br />

Antiretroviral Therapies<br />

December 2008<br />

The Caribbean<br />

The focus of HIV DART 2008 is to assemble<br />

clinicians, researchers, and basic scientists<br />

together to advance our knowledge of the<br />

ongoing drug development processes in<br />

antiretroviral research.<br />

2009<br />

HEP DART 2009<br />

Frontiers in Drug Development for Viral<br />

Hepatitis<br />

December 13-17, 2009<br />

Kohala Coast, Hawaii<br />

The focus of HEP DART 2009 is to assemble<br />

clinicians, researchers, and basic scientists<br />

together to advance our knowledge of the<br />

ongoing drug development processes in the<br />

treatment of hepatitis B and hepatitis C.<br />

1631 Phoenix Boulevard, Suite 4<br />

College Park, GA 30349 USA<br />

Phone: +1 770 997 2484<br />

Fax: +1 770 997 2488<br />

info@informedhorizons.com<br />

www.informedhorizons.com<br />

For more information visit our website at www.informedhorizons.com

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!