Sie sind auf Seite 1von 266

Global Collaboration and Coordination to Advance HIV Vaccine Research

1215 September 2011

Abstract Book

Ministry of Public Health, Thailand

PROGRAM AT A GLANCE
Wednesday, 14 September
Ballroom Foyer | 08:00 18:30 Media Room | 07:00 17:00

AIDS Vaccine 2011


Thursday, 15 September
Ballroom Foyer | 08:00 13:30 Media Room | 07:00 12:00

Monday, 12 September
Registration Speaker Check-In S05 Systems Biology Speaker Check-In Registration Registration Speaker Check-In PL02 New Prevention Strategies Opening Ceremony
Helen Weiss Carolyn Williamson Lynne Mofenson

Tuesday, 13 September

Ballroom Foyer | 08:00 18:00 Media Room | 07:00 17:00

Registration

Ballroom Foyer | 08:00 17:00

Media Room | 12:00 16:00

Speaker Check-In

08:30
S06 New Concepts in Protection Against HIV Acquisitions

Poster Set-Up

08:30

Ballroom B 08:00 12:00

09:00
Ballroom A 08:30 10:00 Ballroom A 08:30 10:00

Presided over by HRH Princess Maha Chakri Sirindhorn Carl Dieffenbach

S07 Characterization of Breakthrough Viruses

09:00

10:00
Tea and Coffee Break
Ballroom B 10:00 10:30 Ballroom A 08:30 10:30 Ballroom B 10:00 10:30

Tea and Coffee Break


Lotus 1 4 08:30 10:30 Lotus 5 7 08:30 10:30

10:00
Tea and Coffee Break
Ballroom B 10:30 11:00

10:30
Pontiano Kaleebu Punnee Pitisuttithum Jerome Kim Barton Haynes Giuseppe Pantaleo

PL01 Novel Approaches in Clinical Evaluation Through Global Collaboration S01 Bridging and Sustaining Community Partners in AIDS Vaccine Research OA07 B Cell Immunology OA08 Animal Models and HIV Transmission

10:30 11:00

PL03 Novel Immunogens and Vaccine Delivery Strategies: Where Do We Go next?


Jeffrey Boyington C. Richter King Steven Reed

Satellite Sessions
Ballroom A 10:30 12:30 Ballroom A 10:30 12:30 Lotus 1 4 10:30 12:30 Lotus 5 7 10:30 12:30

12:30
Ballroom B 12:30 13:30 Ballroom B 12:30 13:30

08:30 15:30 Detailed descriptions are located in the program book.

Ballroom A 11:00 12:30

Lunch Buffet Young and Early-Career Investigators Networking Lunch

Lunch Buffet Young and Early-Career Investigators Networking Lunch

Closing and Handover


Pratap Singhasivanon Dan Barouch Ballroom A 12:30 13:00

12:30

13:30
OA02 Innate Immunity
Lotus 1 4 13:30 15:00 Lotus 5 7 13:30 15:00 Ballroom A 13:30 15:00 Lotus 1 4 13:30 15:00

OA01 Vaccines and Other Prevention Strategies

OA03 Novel Immunogens and Inserts

0A09 Pre-Clinical and Early Clinical Trials

OA10 B Cell Vaccine Concepts

OA11 Muscosal Immunity


Lotus 5 7 13:30 15:00

13:30

Ballroom A 13:30 15:00

15:00
Tea and Coffee Break
Ballroom B 15:00 15:30

Poster Viewing with Tea and Coffee Break


Ballroom B 15:00 16:00

15:00

15:30
OA05 Acute Infection/ Viral Diversity OA06 T-Cell Immunity and Immune Escape

16:00

Pratap Singhasivanon Jos Esparza Stanley Plotkin Sanjay Gurunathan Lotus 1 4 15:30 17:00 Lotus 5 7 15:30 17:00

Special Session

OA04 Novel Monoclonals and Structural Insights

Ballroom A 16:00 17:10

Ballroom A 15:30 17:15

S02 Recent Advances in B Cell & Protective Antibody Responses


Ballroom A 16:00 18:00

S03 The Role of Host Genetics in Innate and Adaptive Immunity


Lotus 1 4 16:00 18:00

S04 Integrated an Emerging Novel Issue in Clinical Trials


Lotus 5 7 16:00 18:00

16:00

17:10

Poster Session 01* and Welcome Reception

17:15
Poster Session 02* and Reception
*All posters ending in even numbers will be presented. Ballroom B 18:00 19:30
All posters must be removed at the conclusion of Poster Session 02. Remaining posters will be discarded by the convention center staff.

*All posters ending in odd numbers will be presented.

Banquet at the Royal Navy Hall

18:00

Ballroom B 17:10 18:40

Buses will depart from the Centara Grand and Holiday Inn hotels at 17:15. Buses will depart the Navy Hall to return to the Centara and Holiday Inn hotels at 21:00.

Poster Dismantle

Abstracts presented at

AIDS

Vaccine 2011
1215 September 2011
Patumwan, Bangkok, Thailand
www.VaccineEnterprise.org www.mahidol.ac.th http://eng.moph.go.th

Abstracts will be published as an online supplement in AIDS Research and Human Retroviruses. Abstracts will be open access at http://www.liebertonline.com/aid at the conclusion of the conference on Thursday, 15 September.

AIDS Vaccine 2011 Partners

www.anrs.fr

www.gatesfoundation.org

www.chinacdc.cn/en

www.chvi-icvv.gc.ca/new-eng.html +

www.pedaids.org

www.geovax.com

www.gsk.com

www.iavi.org

www.mabtech.com

www.niaid.nih.gov

www.nstda.or.th/eng

www.oar.nih.gov

www.sanofipasteur.com

www.aacvb.org/members/TCEB.html

www.mfa.go.th/web/2201.php

www.tourismthailand.org

www.unaids.org

www.hivresearch.org

www.wellcome.ac.uk

www.xstrataalloys.com

The views expressed in written conference materials or publications and by speakers and moderators at HHS-sponsored conferences do not necessarily reflect the official policies of the Department of Health and Human Services (HHS), nor does mention of trade names, commercial practices, or organizations imply endorsement by the U.S. Government. + Production of this abstract book has been made possible through a financial contribution from the Public Health Agency of Canada. The views expressed herein do not necessarily represent the views of the Public Health Agency of Canada.

AIDS Vaccine 2011

Contents
Program at a Glance . . . . . . . . . . . . . . . . . . . Inside Front Cover Conference Organizers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . iivi Award and Scholarship Recipients . . . . . . . . . . . . . . . . . . viixi Program Monday, 12 September . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 Tuesday, 13 September . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27 Wednesday, 14 September . . . . . . . . . . . . . . . . . . . . . . . . . 815 Thursday, 15 September . . . . . . . . . . . . . . . . . . . . . . . . . . 1618 Presentations Special Session . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 Opening Ceremony . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 Plenary Sessions 0103 . . . . . . . . . . . . . . . . . . . . . . . . . . 2127 Symposia 0107 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2848 Oral Abstract Sessions 0111 . . . . . . . . . . . . . . . . . . . . . . 4984 Posters 0121 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85230 Author Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231239 Notes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 240246 Certificate of Attendance . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
ABBREVIATIONS
Late Breaker. . . . . . . . . . . . . . . . . LB Posters . . . . . . . . . . . . . . . . . . . . . . P Oral Abstract. . . . . . . . . . . . . . . . OA Symposia. . . . . . . . . . . . . . . . . . . . S Opening Ceremony . . . . . . . . . . OC Special Session. . . . . . . . . . . . . . SS Plenary Sessions. . . . . . . . . . . . . PL

S AV E T H E D AT E

912 SEPTEMBER 2012


BOSTON, MASSACHUSETTS, USA
AIDS Vaccine 2011 i

Conference Organizers
Chair
Pratap Singhasivanon

CONFERENCE ORGANIZERS

Faculty of Tropical Medicine, Mahidol University, Thailand

Co-Chairs
Punnee Pitisuttithum
Faculty of Tropical Medicine, Mahidol University, Thailand

Manit Teeratantikanont

Department of Disease Control, Ministry of Public Health, Thailand

Local Organizing Committee


Somsak Akksilp
Ministry of Public Health, Thailand

Charles Pamonmontri Praphan Phanuphak

Ministry of Public Health, Thailand Thai Red Cross AIDS Research Centre, Thailand

Supachart Bandhumani

Immigration Bureau of Thailand, Thailand

Chamaiporn Bumrungsin

Landside Operations Department, Suvarnabhumi Airport, Airports of Thailand Public Company Limited (AOT), Thailand

Yaowapa Pratoomsuwan

Faculty of Tropical Medicine, Mahidol University, Thailand

Thanaporn Chaibongsai Panada Chanitwat Vichai Chokevivat

Sasithon Pukrittayakamee Supachai Rerks-Ngarm Kiat Ruxrungtham

Thai Airways International Public Company Limited, Thailand Thai Airways International Public Company Limited, Thailand Government Pharmaceutical Organization (GPO), Thailand

Faculty of Tropical Medicine, Mahidol University, Thailand CDC Department, Ministry of Public Health, Thailand Division of Allergy and Clinical Immunology, Faculty of Medicine, Chulalongkorn University, Thailand

Kachit Choopanya

Bangkok Tenofovir Study Group (BTSG), Bangkok Metropolitan Administration (BMA), Thailand

Thira Sirisanthana

Research Institute for Health Sciences, Chiang Mai University, Thailand

Supamit Chunsuttiwat Pokrath Hansasuta Tamashiro Hiko

Sugree Sithivanich

Ministry of Public Health, Thailand Faculty of Medicine, Chulalongkorn University, Thailand Graduate School of Medicine, Department of Global Health and Epidemiology (GHE), Hokkaido University, Japan

Tourism Authority of Thailand (TAT), Thailand

Vichaya Soonthornsaratoon Ruengpung Sutthent

Thailand Convention and Exhibition Bureau, Thailand Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand

Amorn Leelarasamee

Thitika Teeranetr

Division of Infectious Disease and Tropical Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand

Faculty of Tropical Medicine, Mahidol University, Thailand

Prasert Thongcharoen Sirisak Warintarawat Chantapong Wasi

Suprabha Moleeratanond Viseth Ngauy

Virology Association, Thailand Ministry of Public Health, Thailand Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand

Thailand Convention and Exhibition Bureau, Thailand Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand

Sorachai Nitayaphan

Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand

Scientific Coordinator Srisin Khusmith

Faculty of Tropical Medicine, Mahidol University, Thailand

Secretary to the Local Organizing Committee Paul Adams

Faculty of Tropical Medicine, Mahidol University, Thailand

Communications and Media Relations Coordinator Kasinee Buchachart


Faculty of Tropical Medicine, Mahidol University, Thailand

Assistant Secretary to the Local Organizing Committee Pornpimon Adams


Faculty of Tropical Medicine, Mahidol University, Thailand

Logistics Coordinator Jitra Waikagul

Faculty of Tropical Medicine, Mahidol University, Thailand

ii

AIDS Vaccine 2011

Conference Organizers
Consultants to the Local Organizing Committee
Thai Airways International Public Company Limited, Thailand President, Mahidol University, Thailand

Wiboon Bangtamai

Commissioner of the Immigration Bureau, Thailand

Suraphon Svetasreni

Tourism Authority of Thailand (TAT), Thailand

Ekalarp Rattanaruja

Suvarnabhumi Airport Passenger Control Customs Bureau, Thailand

Nirandra Theeranartsin

Airports of Thailand Public Company Limited (AOT), Thailand

Scientific Organizing Committee


Galit Alter
Ragon Institute of MGH, MIT and Harvard, USA

Jerome Kim

Walter Reed Army Institute of Research (WRAIR), USA

Viktor Appay

French National Institute of Health and Medical Research (INSERM), France

Wayne Koff

International AIDS Vaccine Initiative (IAVI), USA

Susan Barnett Dan Barouch

Novartis Vaccines and Diagnostics, Inc., USA Beth Israel Deaconess Medical Center, USA

Roger Le Grand

French Alternative Energies and Atomic Energy Commission (CEA), France

Amapola Manrique John Mascola

Global HIV Vaccine Enterprise, USA Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Alan Bernstein

Global HIV Vaccine Enterprise, USA

Susan Buchbinder

San Francisco Department of Public Health, USA

Bonnie Mathieson

Damien Chaussabel Daniel Douek

Office of AIDS Research, National Institutes of Health, USA

Baylor Institute for Immunology Research, USA Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Francine McCutchan Lynn Morris Gary Nabel

The Bill & Melinda Gates Foundation, USA National Institute for Communicable Diseases, South Africa Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Jacques Fellay Keith Fowke

Duke University, USA University of Manitoba, Canada

Punnee Pitisuttithum Morgane Rolland

Genoveffa Franchini Barney Graham

Faculty of Tropical Medicine, Mahidol University, Thailand University of Washington, USA

National Cancer Institute, National Institutes of Health, USA Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Kiat Ruxrungtham

Glenda Gray

Division of Allergy and Clinical Immunology, Faculty of Medicine, Chulalongkorn University, Thailand

Perinatal HIV Research Unit, South Africa

Jeffrey Safrit

Catherine Hankins Barton Haynes Shiu-Lok Hu

Elizabeth Glaser Pediatric AIDS Foundation, USA

Joint United Nations Programme on HIV/AIDS (UNAIDS), Switzerland Human Vaccine Institute, Duke University, USA University of Washington, USA

Eric Sandstrm Yiming Shao

Karolinska Institutet, Sweden Chinese Center for Disease Control and Prevention, China

Robin Shattock

Margaret Johnston

St Georges, University of London, UK

National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Ruengpung Sutthent

Stephen Kent

Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand

The University of Melbourne, Australia

Steven Wakefield

Srisin Khusmith

HIV Vaccine Trials Network (HVTN), USA

Faculty of Tropical Medicine, Mahidol University, Thailand

AIDS Vaccine 2011

iii

CONFERENCE ORGANIZERS

Piyasvasti Amranand

Piyasakol Sakolsatayadorn

Conference Organizers
AIDS Vaccine Steering Group
Galit Alter
Ragon Institute of MGH, MIT and Harvard, USA

Yves Lvy

CONFERENCE ORGANIZERS

French National Agency for Research on AIDS and Viral Hepatitis, France

Dan Barouch

Beth Israel Deaconess Medical Center, USA

Siobhan Malone

The Bill & Melinda Gates Foundation, USA

Alan Bernstein Jos Esparza

Global HIV Vaccine Enterprise, USA The Bill & Melinda Gates Foundation, USA

Bonnie Mathieson

Office of AIDS Research, National Institutes of Health, USA

Margaret McCluskey Howard Njoo

United States Agency for International Development (USAID), USA Public Health Agency of Canada, Canada

Catherine Hankins Barton Haynes Eric Hunter

Joint United Nations Programme on HIV/AIDS (UNAIDS), Switzerland Human Vaccine Institute, Duke University, USA Center for AIDS Research, Emory University, USA

Punnee Pitisuttithum Rino Rappuoli


Novartis, Italy

Faculty of Tropical Medicine, Mahidol University, Thailand

Pontiano Kaleebu Katherine Kripke

Uganda Virus Research Institute, Uganda National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Pratap Singhasivanon Steven Wakefield

Faculty of Tropical Medicine, Mahidol University, Thailand HIV Vaccine Trials Network (HVTN), USA.

Scientific Review Committee


Alashle Abimiku Todd Allen Galit Alter
University of Maryland, Biotech Institute, USA Ragon Institute of MGH, MIT and Harvard, USA Ragon Institute of MGH, MIT and Harvard, USA

Cynthia Derdeyn Daniel Douek

Emory University, USA Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Jacques Fellay Keith Fowke

Marcus Altfeld

Duke University, USA University of Manitoba, Canada

Ragon Institute of MGH, MIT and Harvard, USA

Jintanat Ananworanich Viktor Appay Dan Barouch

South East Asia Research Collaboration with Hawaii, Thailand French National Institute of Health and Medical Research (INSERM), France Beth Israel Deaconess Medical Center, USA

Genoveffa Franchini Jonathan Fuchs Paul Goepfert

National Cancer Institute, National Institutes of Health, USA San Francisco Department of Public Health, USA University of Alabama at Birmingham, USA

Susan Buchbinder Luigi Buonaguro Dennis Burton

San Francisco Department of Public Health, USA Istituto Nazionale Tumori, Italy The Scripps Research Institute, USA

Barney Graham

Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Glenda Gray

Perinatal HIV Research Unit, South Africa

Damien Chaussabel Robert Chen Joseph Chiu

Tomas Hanke

Baylor Institute for Immunology Research, USA Centers for Disease Control and Prevention, USA Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand

University of Oxford, UK

Alexandre Harari Barton Haynes Shiu-Lok Hu Eric Hunter

Centre Hospitalier Universitaire Vaudois, Switzerland Human Vaccine Institute, Duke University, USA University of Washington, USA Center for AIDS Research, Emory University, USA

Josephine Cox

International AIDS Vaccine Initiative (IAVI), USA

Mark de Souza Patrice Debre

Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand Hopital Pitie Salpetriere, France

Margaret Johnston

National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

iv

AIDS Vaccine 2011

Conference Organizers
Scientific Review Committee (cont.)
The University of Melbourne, Australia The Bill & Melinda Gates Foundation, USA

Srisin Khusmith Jerome Kim

Faculty of Tropical Medicine, Mahidol University, Thailand Walter Reed Army Institute of Research (WRAIR), USA

Kiat Ruxrungtham

Division of Allergy and Clinical Immunology, Faculty of Medicine, Chulalongkorn University, Thailand

Jeffrey Safrit

Wayne Koff

Elizabeth Glaser Pediatric AIDS Foundation, USA

International AIDS Vaccine Initiative (IAVI), USA

Eric Sandstrm

Roger Le Grand Yves Lvy

Karolinska Institutet, Sweden

French Alternative Energies and Atomic Energy Commission (CEA), France French National Agency for Research on AIDS and Viral Hepatitis, France

Gabriella Scarlatti

San Raffaele Scientific Institute, Italy

Alexandra Schuetz Olivier Schwartz Yiming Shao

Amapola Manrique John Mascola

Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand Institut Pasteur, France Chinese Center for Disease Control and Prevention, China

Global HIV Vaccine Enterprise, USA Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Bonnie Mathieson

Office of AIDS Research, National Institutes of Health, USA

Robin Shattock

St Georges, University of London, UK

Margaret McElrath James McIntyre

HIV Vaccine Trials Network (HVTN), USA Anova Health Institute, South Africa

Ruengpung Sutthent

Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand

Ronald Swanstrom Hiko Tamashiro Jim Tartaglia

Andrew McMichael Nelson Michael

University of North Carolina, USA World Health Organization, Switzerland sanofi pasteur, USA

Weatherall Institute of Molecular Medicine, UK U.S. Military HIV Research Program, USA

David Montefiori Lynn Morris Gary Nabel

Duke University, USA National Institute for Communicable Diseases, South Africa Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, USA

Amalio Telenti

University Hospital and University of Lausanne, Switzerland

Caroline Tiemessen Georgia Tomaras

National Institute for Communicable Diseases, South Africa Duke University, USA

Viseth Ngauy

Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand

Alexandra Trkola Yegor Voronin

Institute of Medical Virology, University of Zurich, Switzerland Global HIV Vaccine Enterprise, USA

Sorachai Nitayaphan Jo-Ann Passmore

Armed Forces Research Institute of Medical Sciences (AFRIMS), Thailand University of Cape Town, South Africa

Steven Wakefield

HIV Vaccine Trials Network (HVTN), USA

Praphan Phanuphak

Thai Red Cross AIDS Research Centre, Thailand

Anna-Lise Williamson

Punnee Pitisuttithum

Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, South Africa

Faculty of Tropical Medicine, Mahidol University, Thailand

Susan Zolla-Pazner

Supachai Rerks-Ngarm Morgane Rolland

CDC Department, Ministry of Public Health, Thailand University of Washington, USA

New York University School of Medicine, Veterans Affairs Medical Center, USA

Michael Zwick

The Scripps Research Institute, USA

AIDS Vaccine 2011

CONFERENCE ORGANIZERS

Stephen Kent

Nina Russell

Conference Organizers
Conference Secretariat
Mark Aurigemma Jennifer Brunet

CONFERENCE ORGANIZERS

Communications Consultant Global HIV Vaccine Enterprise, USA

Amapola Manrique

Global HIV Vaccine Enterprise, USA

Conference Solutions

2545 SW Spring Garden St., Suite 150 Portland, OR 97219 USA AIDSVaccine@ConferenceSolutionsInc.com www.ConferenceSolutionsInc.com

vi

AIDS Vaccine 2011

Award and Scholarship Recipients


2011 New Investigator Awardees
AIDS Vaccine 2011 is proud to announce this years New Investigator Awardees. Muhammed Afolabi Amy Chung
Medical Research Council Laboratories, Gambia The University of Melbourne, Australia

Zaza Ndhlovu Bin Su

Ragon Institute of MGH, MIT and Harvard, USA French National Institute of Health and Medical Research (INSERM) U748, Universit de Strasbourg, France

Miguel Lacerda

University of Cape Town, South Africa

2011 Conference Scholarship Recipients


The scholars listed below applied for and were selected to receive a conference scholarship. The scholarships were made possible by generous donations from our partners and from registration funds. Adedayo Adeyemi Hannah Ajoge
Measure Evaluation, Nigeria Department of Microbiology, Faculty of Science, Ahmadu Bello University, Zaria, Nigeria

Lea Brandt

Statens Serum Institut, Denmark

Sarah Brinckmann Justine Brown

University of Oxford, United Kingdom University of Washington, USA

Tomohiro Akahoshi

Center for AIDS Research, Kumamoto University, Japan

Chanson Brumme Olusegun Busari Sun Caijun

Kabamba Alexandre Rafael Almeida Michael Alpert

BC Centre for Excellence in HIV/AIDS, Canada Federal Medical Centre, Ido-Ekiti, Nigeria Guangzhou Institute of Biomedicine and Health, China

National Institute for Communicable Diseases, South Africa University of So Paulo, Brazil Harvard University, USA

Makhtar Camara

Carmen Alvarez Fernandez

Institut DInvestigacions Biomdiques August Pi I Sunyer (IDIBAPS)/ Fundaci Clnic per a la Recerca Biomdica (FCRB), Spain

Laboratory of Bacteriology Virology, Aristide Le Dantec University Hospital, Senegal

Ann Carias

Erica Andersen-Nissen Raiees Andrabi

Northwestern University, USA

University of Washington/Fred Hutchinson Cancer Research Center, USA All India Institute of Medical Sciences, India

Bimal Chakrabarti

International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center/The Scripps Research Institute, USA

Hodei Arberas Eguiluz

Stephane Champiat

Institut DInvestigacions Biomdiques August Pi I Sunyer (IDIBAPS)/ Fundaci Clnic per a la Recerca Biomdica (FCRB), Spain

University of California, San Francisco, France

Rosamund Chapman Gerald Chege

Panagiotis Athanasopoulos Alejandro Balazs Dion Benetatos Alfred Bere

University of Cape Town, South Africa University of Cape Town, South Africa

Royal Holloway University of London, United Kingdom California Institute of Technology, USA Desmond Tutu HIV Foundation, South Africa University of Cape Town, South Africa

Shivan Chetty

University of Cape Town, South Africa

Fernanda Crtes Valerie Cortez

Instituto Oswaldo Cruz/FIOCRUZ, Brazil University of Washington/Fred Hutchinson Cancer Research Center, USA

Jinal Bhiman

National Institute for Communicable Diseases, South Africa

Alberto Crespo Guardo

Marina Biedma

French National Institute of Health and Medical Research (INSERM) U748, Universit de Strasbourg, France

Institut DInvestigacions Biomdiques August Pi I Sunyer (IDIBAPS)/ Fundaci Clnic per a la Recerca Biomdica (FCRB), Spain

Shayesta Dhalla

James Billingsley

University of British Columbia, School of Population and Public Health, Canada

Harvard Medical School, New England Primate Research Center, USA

Christian Boutwell Emma Bowles

Zelda Euler

Academic Medical Center, University of Amsterdam, Netherlands

Ragon Institute of MGH, MIT and Harvard, USA University of Oxford, United Kingdom

Juliana Falivene

Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Argentina

AIDS Vaccine 2011

vii

SCHOLARSHIP / AWARD RECIPIENTS

Award and Scholarship Recipients


2011 Conference Scholarship Recipients (cont.)
Emilia Falkowska
The Scripps Research Institute, USA

Lenine Liebenberg

University of Cape Town Medical School, South Africa

Anne-Laure Flamar Ana Gervassi

Baylor Institute for Immunology Research, USA Seattle Biomedical Research Institute, USA

Madelene Lindqvist Cynthia Maeto

Ragon Institute of MGH, MIT and Harvard, USA Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Argentina

Shari Gordon

National Cancer Institute, National Institutes of Health, USA

Alison Mahan

Francisco Guenaga

Ragon Institute of MGH, MIT and Harvard, USA

International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center, USA

Sharmila Mohana Rama Reddy Adi Moseri Kim Mous

The University of Melbourne, Australia Weizmann Institute of Science, Israel University of Antwerp, Belgium

Xuan He

Chinese Center for Disease Control and Prevention, China

SCHOLARSHIP / AWARD RECIPIENTS

Tomer Hertz

University of Washington/Fred Hutchinson Cancer Research Center, USA

Henoch Hong Jue Hou

Harvard Medical School, USA Chinese Center for Disease Control and Prevention, China

Boris Murashev Megan Murphy

State Research Institute of Highly Pure Biopreparations, Russian Federation Emory University, USA

Natalie Hutnick Rajesh Jacob

University of Pennsylvania, USA Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, South Africa

Karuppiah Muthumani Roman Ntare

University of Pennsylvania School of Medicine, USA University of Cape Town, South Africa

Rashmi Jalah

Peter Oballah

National Cancer Institute at Frederick, USA

Makerere University Walter Reed Project, Uganda

Gabriela Junqueira Calazans Yuanxi Kang

Irene Onyango

Centro de Referncia e Treinamento DST/AIDS, Brazil AIDS Institute, LKS Faculty of Medicine, The University of Hong Kong, China

Kenya AIDS Control Project (University of Nairobi and University of Manitoba), Kenya

Alexandra Ortiz Craig Pace

University of Pennsylvania, USA Aaron Diamond AIDS Research Center, USA

Thomas Kerkhof

Academic Medical Center, University of Amsterdam, Netherlands

Freddie Mukasa Kibengo Nichole Klatt

Medical Research Council/Uganda Virus Research Institute, Uganda National Institutes of Health/National Institutes of Allergy and Infectious Diseases/Laboratory for Molecular Medicine, USA

Maria de los Angeles Pando

Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Argentina

Vincent Pavot

Alexander Kliche Pratima Kunwar

Institut de Biologie et Chimie des Protines, Centre National de la Recherche Scientifique, France

University of Regensburg, Germany University of Washington, USA

Maria Paximadis Anabela Picton

National Institute for Communicable Diseases, South Africa National Institute for Communicable Diseases/University of the Witwatersrand, South Africa

Alexandre Lederle

French National Institute of Health and Medical Research (INSERM) U748, Universit de Strasbourg, France

Roger Keith Reeves Susan Ribeiro

Mopo Radebe Leshwedi Dan Li

Harvard Medical School, New England Primate Research Center, USA University of So Paulo, Brazil

University of KwaZulu Natal, South Africa Chinese Center for Disease Control and Prevention, China

Catherine Riou

Hualin Li

Beth Israel Deaconess Medical Center, USA

Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, South Africa

Yuxing Li

Raabya Rossenkhan Jonah Sacha

International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center/The Scripps Research Institute, USA

University of Botswana/Botswana Harvard AIDS Institute Partnership, USA Oregon Health & Science University, USA

viii

AIDS Vaccine 2011

Award and Scholarship Recipients


2011 Conference Scholarship Recipients (cont.)
Kotou Sangare
GAIA Vaccine Foundation, Mali

Marit van Gils

Academic Medical Center, University of Amsterdam, Netherlands

Daniel Sheward

University of Cape Town, South Africa

Griet Van Roey

St Georges, University of London, UK

Simon Sigirenda Anthony Smith Kellie Smith

Uganda Virus Research Institute, Uganda University of Minnesota, USA University of Pittsburgh School of Medicine, USA

Sundaram Vishwanathan Yanmin Wan Yaoyu Wang

Centers for Disease Control and Prevention, USA Shanghai Public Health Clinical Center, China Dana-Farber Cancer Institute, USA

Damien Soghoian Ali Ssetaala

Ragon Institute of MGH, MIT and Harvard, USA Uganda Virus Research Institute/International AIDS Vaccine Initiative/ HIV Vaccine Program, Uganda

Constantinos Wibmer Leia Wren

National Institute for Communicable Diseases, South Africa The University of Melbourne, Australia

Kathryn Stephenson Ingemar Sundling Leandro Tarosso

Tian Xiaoling

Beth Israel Deaconess Medical Center, USA Karolinska Institutet, Sweden University of So Paulo, Brazil

Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, China

Lifei Yang

Institut Pasteur of Shanghai, Chinese Academy of Science, China

Tingting Yuan Anli Zhang

AIDS Institute, The University of Hong Kong, China Institutes of Biomedical Sciences, Fudan University, China

Christina Thobakgale Gabriela Turk

HIV Pathogenesis Programme, University of KwaZulu-Natal, South Africa Argentinean National Reference Center for AIDS, Argentina

Mei-Yun Zhang Mingkui Zhou

The University of Hong Kong, China Georg-Speyer-Haus Institute for Biomedical Research, Germany

Nicolas Vabret

Institut Pasteur, France

Attendee Scholarships
Mahidol University provided scholarships for the following conference attendees. James Fletcher Yan Liu
SEARCH/The Thai Red Cross AIDS Research Centre, Thailand Hokkaido University, Japan

Sunee Sirivichayakul

Chulalongkorn University, Thailand

Nopporn Sittisombut Hukumraj Thapa

Faculty of Medicine, Chiang Mai University, Thailand Friends of Hope, Nepal

Angsana Phuphuakrat

Faculty of Medicine, Ramathibodi Hospital, Thailand

The following scholarships to South African HIV vaccine trial site community representatives were made possible by generous support from Xstrata Alloys, the largest Ferrochrome producer in the world. Babalwa Foto
Boitekong Health Centre, South Africa

Mildred Busisiwe Msimanga Nhlanhla Herbet Msimanga Pinki Pooe

Community Member, South Africa Bosabosele Primary School, South Africa Municipality, South Africa

Benjamin Makhoana Nkosinathi Mngadi

The Aurum Institute, South Africa The Aurum Institute, South Africa

Welheminah Kgomotso Moeng


Rustenburg Hospice, South Africa

Kedibone Setsepu

Wilmed Park Hospital, South Africa

George Moteothata Mokhutswane


Department of Health, South Africa

Samuel Tshabangu

The Aurum Institute, South Africa

AIDS Vaccine 2011

ix

SCHOLARSHIP / AWARD RECIPIENTS

Award and Scholarship Recipients


Attendee Scholarships (cont.)
The Thailand International Development Cooperation Agency (TICA) provided scholarships for the following conference attendees. Budiman Bela
Department of Microbiology, University of Indonesia, Indonesia

Sanjay Mendiratta Chao Qiu

All India Institute, Research Institute for Tropical Medicine, India Fudan University, China

Rossana A Ditangco Mario Antonio II Jiz

Research Institute of Tropical Medicine, Manila, Philippines Immunology Department, Research Institute for Tropical Medicine, Philippines

Rozainanee Mohd Zain

Ministry of Public Health, Malaysia

Complimentary Registration Recipients


Mahidol University provided complimentary registrations to the following conference attendees.

SCHOLARSHIP / AWARD RECIPIENTS

Jotika Boonlong

Thai Food and Drug Administration, Thailand

Nathamon Ngaosuwankul

Faculty of Tropical Medicine, Mahidol University, Thailand

Thitilat Chiraunyanann

Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand

Kamol Phojanamongkolkij Patrawadee Pitakpolrat Pakdee Pothisiri

Somdej Pra Boromrajathevi na Sriracha Hospital, Thailand Faculty of Medicine, Chulalongkorn University, Thailand Office of the National Anti-Corruption Commission, Thailand

Anupong Chitwarakorn Pokrath Hansasuta

Department of Disease Control, Ministry of Public Health, Thailand Faculty of Medicine, Chulalongkorn University, Thailand

Navin Horthongkham

Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Thailand

Supachai Sakkhachornphop Satit Sirising

Research Institute for Health Sciences, Chiang Mai University, Thailand Faculty of Science, Mahidol University, Thailand

Prathanporn Kaewpreedee Chutitorn Ketloy

Faculty of Medicine, Chulalongkorn University, Thailand Faculty of Medicine, Chulalongkorn University, Thailand

Ruengpung Sutthent

Faculty of Medicine, Siriraj Hospital, Thailand

Chirasak Khamboonruang

Somsit Tansuphaswadikul Vipa Thanachartwet

Centers for Disease Control and Prevention, Ministry of Public Health, Thailand

Bumrasnaradura Hospital, Thailand Faculty of Tropical Medicine, Mahidol University, Thailand

Doungnapa Kingkeow Yupin Lawanprasert

Research Institute for Health Sciences, Chiang Mai University, Thailand Thai Food and Drug Administration, Thailand

Pattarawat Thantiworasit LTC Kwanjai Viputtigul

Faculty of Medicine, Chulalongkorn University, Thailand Armed Forces Research of Medical Science (AFRIMS), Thailand

Bhassorn Limanonda Preeda Malasit

Faculty of Medicine, Chulalongkorn University, Thailand Mahidol University, Thailand

Surasak Wiboonchutikul Jiraprapa Wipasa Napa Wongsilp Suthee Yoksan

Faculty of Medicine, Ramathibodi Hospital, Thailand Research Institute for Health Sciences, Chiang Mai University, Thailand Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand Institute of Molecular Biosciences, Mahidol University, Thailand

Kumthorn Malathum

Faculty of Medicine, Ramathibodi Hospital, Thailand

Weerawat Manosuthi

Bumrasnaradura Hospital, Thailand

AIDS Vaccine 2011

Award and Scholarship Recipients


Complimentary Registration Recipients (cont.)
The Armed Forces Research Institute of Medical Sciences (AFRIMS) provided complimentary registrations to the following conference attendees. Prasert Auewarakul
Siriraj Hospital, Thailand

Anchana Palasudhi Krit Palasudhi

Ministry of Public Health, Thailand Ministry of Public Health, Thailand

Chureeratana Bowonwatanuwong
Chonburi Hospital, Thailand

Sasisopin Kiertiburanakul Suda Louisirirotchanakul


Siriraj Hospital, Thailand

Faculty of Medicine, Ramathibodi Hospital, Thailand

Nakorn Premsri

Centers for Disease Control and Prevention, Ministry of Public Health, Thailand

Boonmee Sathapatayavongs Somnuek Sungkanuparph

Chawetsan Namwat

Faculty of Medicine, Ramathibodi Hospital, Thailand Faculty of Medicine, Ramathibodi Hospital, Thailand

The Department of Disease Control, Ministry of Public Health, Thailand, provided complimentary registrations to the following conference attendees. Somyot Kittimunkong
Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand

Petchsri Sirinirund Usa Thisyakorn

Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand

Attaya Limwattanayingyong Pairoj Saonuam

Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand Bureau of AIDS,TB and STIs, Ministry of Public Health, Thailand

The National Science and Technology Development Agency (NSTDA), Thailand, provided complimentary registrations to the following conference attendees. Akachai Prompet Pinya Pulsawat
Faculty of Medicine, Chulalongkorn University, Thailand Faculty of Medicine, Chulalongkorn University, Thailand

Yada Tansiri

Faculty of Medicine, Chulalongkorn University, Thailand

Hathairat Thananchai

Chiang Mai Medical School, Thailand

AIDS Vaccine 2011

xi

SCHOLARSHIP / AWARD RECIPIENTS

Centers for Disease Control and Prevention, Ministry of Public Health, Thailand

xii

AIDS Vaccine 2011

Monday, 12 September

Program
Ballroom Foyer, Level 22 Media Room, Level 22 Ballroom A, Level 22

Monday, 12 September
08:00 18:00 12:00 16:00 16:00 17:10 16:00 16:10 Registration Speaker Check-In

Special Session
Chairs: Punnee Pitisuttithum and Manit Teeratantikanont Pratap Singhasivanon 2011 Conference Chair Faculty of Tropical Medicine, Mahidol University Bangkok, Thailand Jos Esparza Global HIV Vaccine Enterprise Acting President of the Board Bill & Melinda Gates Foundation Seattle, WA, USA

16:10 16:20

16:20 16:40

Are There Lessons from Past Vaccines for an AIDS Vaccine? SS01.01 Stanley Plotkin University of Pennsylvania Doylestown, PA, USA Moving Forward from RV144: An Industrial Perspective SS01.02 Sanjay Gurunathan sanofi pasteur Swiftwater, PA, USA

16:40 17:10

17:10 18:40

All posters ending in odd numbers will be presented during Poster Session 01. Presenting authors will be available for discussion at this time.

AIDS Vaccine 2011

PROGRAM / MONDAY

Poster Session 01 and Welcome Reception

Ballroom B, Level 22

Program
Tuesday, 13 September
08:00 17:00 07:00 17:00 08:30 10:00 08:30 08:45 08:45 09:00 09:20 Registration Speaker Check-In

Tuesday, 13 September

Grand Ballroom Foyer, Level 22 Media Room, Level 22 Ballroom A, Level 22

Opening Ceremony
Doors Open. Entertainment Provided by Mahidol University Orchestra Doors Close. All guests must be seated for the arrival of Her Royal Highness. Opening Remarks Her Royal Highness Princess Maha Chakri Sirindhorn

09:20 09:50

Progress Towards an HIV Vaccine Lessons Learned from HIV Prevention OC01.01 Carl Dieffenbach Division of AIDS, NIAID, NIH Bethesda, MD, USA Tea and Coffee Break Ballroom B, Level 22

10:00 10:30

10:30 12:30

Plenary Session 01:

Novel Approaches in Clinical Evaluation through Global Collaboration


Chairs: Nelson Michael and Punnee Pitisuttithum

Ballroom A, Level 22

10:30 10:45

Africas Contribution to AIDS Vaccine Research and Development PL01.01 Pontiano Kaleebu MRC/UVRI Research Unit on AIDS Entebbe, Uganda Thailands Contribution to HIV Vaccine Research and Development PL01.02 Punnee Pitisuttithum Faculty of Tropical Medicine, Mahidol University Bangkok, Thailand Update on RV144: Correlates, Sieve Analysis, and Clinical Development PL01.03 Jerome Kim Walter Reed Army Institute of Research (WRAIR) Rockville, MD, USA Case Control Study of the RV144 Trial for Immune Correlates: PL01.04 The Analysis and Way Forward Barton Haynes Duke Human Vaccine Institute Durham, NC, USA Poxvirus Vector-Based HIV Vaccines Beyond RV144 PL01.05 Giuseppe Pantaleo Centre Hospitalier Universitaire Vaudois (CHUV) Lausanne, Switzerland

10:45 11:00

11:00 11:30

11:30 12:00 PROGRAM / TUESDAY

12:00 12:30

AIDS Vaccine 2011

Tuesday, 13 September
12:30 13:30 Lunch Buffet Young and Early-Career Investigators Networking Lunch

Program
Ballroom B, Level 22

Lunch tables in Ballroom B have been reserved for young and early-career investigators to network with senior researchers. A list of featured researchers is available at the registration desk.

13:30 15:00 13:30 13:45

Oral Abstract Session 01:

Vaccines and Other Prevention Strategies


Chairs: Chris Beyrer and Glenda Gray

Ballroom A, Level 22

DNA/NYVAC Vaccine Regimen Induces HIV-Specific CD4 and OA01.01 CD8 T-Cell Responses in Intestinal Mucosa M Perreau, HC Welles, A Harari, O Hall, R Martin, M Maillard, G Dorta, P Bart, EJ Kremer, J Tartaglia, R Wagner, M Esteban, Y Levy, G Pantaleo Cell-Mediated Immune Responses After DNA Delivered by Either OA01.02 Biojector or Electroporation and Boosted with a Heterologous Insert Recombinant Poxvirus JR Currier, S Ratto-Kim, V Ngauy, J Ake, C Lau, T Doris, E Herrera, WB David, NY Sardesai, J Boyer, AS Khan, J Lee, M Bagarazzi, J Yan, E Adams, P Earl, B Moss, J Kim, N Michael, M Robb, MA Marovich Titered Mucosal Challenge of Rhesus Macaques with SIVmac251 OA01.03 Recapitulates HIV Vaccine Efficacy in Humans G Franchini, P Pegu, M Vaccari, S Gordon, B Keele, M Doster, Y Guan, G Ferrari, D Montefiori, D Venzon, C Fenizia, J Lifson, N Michael, J Kim, J Tartaglia Preservation of HIV-1-Specific CD4+ T-IFN-Cell Responses in OA01.04 Intercurrent Infections Following Exposure to Tenofovir Gel M Mureithi , D Poole, V Naranbhai, S Reddy, Np Mkhwanazi, S Sibeko, Q Abdool Karim, S abdool Karim, T Ndungu, M Altfeld, CAPRISA 004 Trial Group Homologous Priming and Boosting with Recombinant MVA, OA01.05 Prevention of Infection as Good as With the Heterologous Regimen of DNA Priming and MVA Boosting HL Robinson, L Lai, S Kwa, PA Kozlowski, V Hirsch, BK Felber, GN Pavlakis, PL Earl, B Moss, RR Amara Extended Evaluation of Volunteers Who Become HIV-1 Infected During Participation in a Phase III Vaccine Trial of ALVAC-HIV and AIDSVAX B/E S Rerks-Ngarm, RM Paris, S Chunsutthiwat, N Premsri, C Namwat, J Bowornwatanuwong, S Li, J Kaewkungkal, R Trichavaroj, M de Souza, D Francis, E Adams, S Gurunathan, J Tartaglia, R OConnell, C Eamsila, S Nitayaphan, V Ngauy, P Thongcharoen, P Kunasol, N Michael, M Robb, P Gilbert, J Kim OA01.06 LB

13:45 14:00

14:00 14:15

14:15 14:30

14:30 14:45

14:45 15:00

AIDS Vaccine 2011

PROGRAM / TUESDAY

Program
13:30 15:00 13:30 13:45

Tuesday, 13 September
Lotus 1 - 4, Level 22

Oral Abstract Session 02:


Innate Immunity
Chairs: Jean-Louis Excler and Thira Sirisanthana

Innate Immune Response Signatures to Ad5 (HVTN 071) and OA02.01 MVA (HVTN 205/908) Vectored Candidate HIV Vaccines Predict Induction of Adaptive Responses E Andersen-Nissen, DE Zak, AC Duerr, JT Chang, ER Peterson, AT Krishnamurty, TR Hensley, DJ Adams, MK Hamilton, J Li, A Sato, CA Morgan, ML Elizaga, SC DeRosa, PA Goepfert, HL Robinson, A Aderem, MJ McElrath Frequent and Strong NK Responses to HIV-1 Env Peptides in Individuals OA02.02 with Acute and Chronic HIV-1 Infection
C.F. Thobakgale, L. Fadda, K. Lane, F. Pereyra, B.D. Walker, T. Allen, M. Altfeld

13:45 14:00

14:00 14:15

NKp46 and NKG2D Receptors Are Implicated in the Priming and OA02.03 Suppression of HIV Replication by Natural Killer Cells Stimulated by MVA/HIV ANRS Vaccine J Cummings, V Arnold, C Didier, A Gilbert, K Yarbrough, Y Levy, F Barr-Sinoussi, D Scott-Algara HIV Infection Eliminates the Antibody-Dependent Cellular OA02.04 Cytotoxicity (ADCC) Functional Advantage of KIR3DL1-Expressing NK Cells from HLA-Bw4 Carriers MS Parsons, L Wren, G Isitman , M Navis, I Stratov, NF Bernard, SJ Kent Inhibition of Human Immunodeficiency Virus Type 1 Infection of OA02.05 Human Monocyte-Derived Macrophages by Anti-Lipid and Anti-MPER Monoclonal Antibodies M Rao, O Jobe, KK Peachman, GR Matyas, LV Asher, CR Alving JAK3 Inhibition In Vivo in Chronically SIV Infected Rhesus Macaques Deplete NK-17 Cells L Kowawisatsut, AA Ansari, K Pattanapanyasat OA02.06 LB

14:15 14:30

14:30 14:45

14:45 15:00

13:30 15:00 13:30 13:45

Oral Abstract Session 03:

Novel Immunogens and Inserts


Chairs: Dan Barouch and Kiat Ruxrungtham

Lotus 5 - 7, Level 22

Targeting Antigen to CD40 on Dendritic Cells Mediates Expansion OA03.01 of Multi-Epitope HIV-Specific Polyfunctional CD4+ and CD8+ T Cells A Flamar, Y Xue, S Zurawski, S Oh, M Montes, A Montes, B King, L Sloan, J Banchereau, Y Levy, G Zurawski Identification of Escape-Refractory Subdominant CD8 Epitopes for OA03.02 Common HLA Alleles CL Boutwell, C Oniangue-Ndza, A Schneidewind, H Streeck, A Seese, E Mellors, K Murthy, K Power, M Kemper, T Dudek, BD Walker, M Altfeld, TM Allen

13:45 14:00 PROGRAM / TUESDAY 4

AIDS Vaccine 2011

Tuesday, 13 September
14:00 14:15

Program

HIV-1 gp120 V1V2-Scaffolds for Structural Analysis and Immunogen Design OA03.03 J McLellan, M Pancera, K Dai, J Boyington, Z Yang, C Carrico, S Shahzad-ul-Hussan, M Sastry, S Schmidt, Y Yang, M Bonsignori, B Haynes, S Phogat, C Bewley, J Mascola, W Schief, G Nabel, P Kwong Identification of Mimotopes for Neutralizing Antibodies from OA03.04 Elite Controllers with Env-Tailored Phage Display Libraries M Zhou, S Koch, T Meyer, B Brill, M Sakarellos-Daitsiotis, Miguel Benito, V Soriano, A Haberl, M Bickel, H von Briesen, M Hust, U Dietrich Role of Hypervariable Loops in Intersubunit Associations of Env: Trimeric gp140 Immunogen as a Platform for Novel Epitope Display H Cheng, CG Moscoso, L Xing, L Martin, S Barnett, I Srivastava OA03.05 LB

14:15 14:30

14:30 14:45

14:45 15:00

Development and Characterization of HIV-1 Virus-like Particles OA03.06 Produced by Stably Transfected Drosophila S2 cells L Yang, Y Song, P Zhu, P Zhou Tea and Coffee Break Ballroom B, Level 22

15:00 15:30

15:30 17:15 15:30 15:45

Oral Abstract Session 04:

Novel Monoclonals and Structural Insights


Chairs: Srisin Khusmith and Peter Kwong

Ballroom A, Level 22

Potent and Broad Neutralization by a CD4 Binding Site OA04.01 Monoclonal Antibody from an HIV-1 Infected Donor E Falkowska, DR Burton, P Poignard, J Mascola, PD Kwong, X Wu Human Anti-V2 Monoclonal Antibodies Neutralize Tier 1 OA04.02 HIV-1 Pseudoviruses MK Gorny, C Williams, T ONeal, X Wang, MS Seaman, S Zolla-Pazner 2F5-like Cross-Reactive HIV-1-Neutralizing Monoclonal Antibodies OA04.03 with Low Number of Somatic Mutations Z Zhu, HR Qin, W Chen, Q Zhao, X Shen, R Schutte, Y Wang, G Ofek, E Streaker, P Prabakaran, GG Fouda, H Liao, J Owens, M Louder, Y Yang, K Klaric, MA Moody, JR Mascola, JK Scott, PD Kwong, D Montefiori, BF Haynes, GD Tomaras, DS Dimitrov Characterization of CD4-Binding Site Directed Monoclonal Antibodies OA04.04 Isolated from HIV-1 gp140 Env Immunized Rhesus Macaques C Sundling, Y Li, N Huynh, R Wilson, S ODell, JR Mascola, RT Wyatt, GB Karlsson Hedestam Structural Insights into HIV-1 Neutralization by New Highly Potent OA04.05 and Broadly Neutralizing Antibodies J Julien, R Pejchal, L Kong, RS Stanfield, J Lai, LM Walker, KJ Doores, M Huber, E Falkowska, P Poignard, SK Phogat, WC Koff, DR Burton, IA Wilson AAV2/8-Encoded Vaccine Expressing a Broadly Neutralizing HIV Antibody OA04.06 Protects Humanized Mice from High-Dose Intravenous Viral Challenge AB Balazs, J Chen, CM Hong, DS Rao, L Yang, D Baltimore Evolution of HIV-1 Transmitted/Founder Viruses Results in the Formation of Epitopes for Later Broadly Rross-Neutralizing Antibodies PL Moore, ES Gray, T Hermanus, D Sheward, N Tumba, CK Wibmer, J Bhiman, S Sibeko, SS Abdool Karim, C Williamson, L Morris OA04.07 LB

15:45 16:00

16:00 16:15

16:15 16:30

16:30 16:45

16:45 17:00

17:00 17:15

AIDS Vaccine 2011

PROGRAM / TUESDAY

Program
15:30 17:00 15:30 15:45

Tuesday, 13 September
Lotus 1 4, Level 22

Oral Abstract Session 05:


Acute Infection/Viral Diversity
Chairs: Jintanat Ananworanich and Deborah Birx

HIV-Specific Cytolytic CD4+ T-Cell Responses During Acute OA05.01 HIV-1 Infection Predict Disease Outcome DZ Soghoian, H Jessen, S Cutler, M Flanders, S Ranasinghe, M Lindqvist, I Davis, K Lane, J Rychert, ES Rosenberg, BD Walker, H Streeck Control of Acute HIV-1 Subtype C Viremia Is Associated with OA05.02 Limited CD8+ T-Cell Immunogenicity MA Radebe, K Nair, F Chonco, K Bishop, K Bishop, JK Wright, M van der Stok, Z Mncube, M Altfeld, BD Walker, T Ndungu Lymphocyte Dynamics in Acute HIV-1 Infection: OA05.03 RV217-The Early Capture HIV Cohort Study (ECHO) L Eller, M Eller, A Eser, P Naluyima, K Shikuku, A Taylor, E Rono, S Sukwit, S Nitayaphan, J Currier, M de Souza, S Peel, J Kim, N Michael, M Marovich, M Robb Long-Lived Historical Record of Evolution of Viral Diversity OA05.04 in Peripheral Blood Cells by Deep Pyrosequencing L Yin, L Liu, Y Sun, W Hou, AC Lowe, M Salemi, WG Farmerie, JW Sleasman, MM Goodenow Dynamics of HIV-1 Subtype C Gag in the Global Epidemic OA05.05 V Novitsky, R Wang, M Essex Important Contribution of Subdominant HIV-Specific CD8 T Cell OA05.06 Responses to Early Control of HIV Replication H Streeck, D Kwon, M Addo, M Flanders, I Davis, S Cutler, J Rychert, K Lane, B Yassine-Diab, J Routy, S Little, H Jessen, A Kelleher, F Hecht, R Sekaly, E Rosenberg, B Walker, T Allen, M Altfeld

15:45 16:00

16:00 16:15

16:15 16:30

16:30 16:45 16:45 17:00

15:30 17:00 15:30 15:45

Oral Abstract Session 06:

T-Cell Immunity and Immune Escape


Chairs: Mark de Souza and Pokrath Hansasuta

Lotus 5 - 7, Level 22

Deconstructing HIV-Specific CD8+T Cell Responses in OA06.01 HIV Elite Controllers ZM Ndhlovu, New Investigator Award Winner LB Chibnik, J Proundfoot, S Vine, F Porichis, A McMullen, K Cesa, A Piechocka-Trocha, PL De Jager, DE Kaufmann, BD Walker Ontogeny of CD8+ T Cells in Acute HIV-1 that Inhibit Autologous OA06.02 and Heterologous Transmitted/Founder (T/F) Viruses SA Freel, RA Picking, G Ferrari, C Ochsenbauer, JC Kappes, J Kirchherr, KJ Weinhold, K Soderberg, AJ McMichael, BF Haynes, GD Tomaras The RV144 Vaccine Induced CD4+ T Cells Producing Th1 and OA06.03 T h2 Cytokines with Proliferative Potential S DeRosa, N Frahm, OD Defawe, D Carter, EP Thomas, R Smith, R Gottardo, N Kochar, H Janes, Y Fong, X Yu, NL Michael, JH Kim, S Rerks-Ngarm, MJ McElrath

15:45 16:00 PROGRAM / TUESDAY

16:00 16:15

AIDS Vaccine 2011

Tuesday, 13 September
16:15 16:30

Program

Association of Timing of HLA B*8101 and B*3910 Mediated p24 OA06.04 Sequence Evolution and Pathways to CTL Escape with Disease Progression RS Ntale, DR Chopera, N Ngandu, D Debra, M Mlotshwa, CM Gray, K Mlisana , S Abdool Karim, C Williamson, The CAPRISA 002 Study Team Evidence of HIV-Specific ADCC Immune Escape OA06.05 AW Chung, New Investigator Award Winner G Isitman, M Navis, M Kramski, RJ Center, SJ Kent, I Stratov Imbalance Between T Helper Type 17 and T Regulatory Cells: OA06.06 Implications for HIV Pathogenesis D Li, J Chen, M Jia, K Hong, Y Ruan, H Liang, H Peng, P Ma, Y Shao

16:30 16:45

16:45 17:00

17:15 21:30 17:15 17:30 18:00 19:00 19:00 21:00 21:00 21:30

Banquet at the Royal Navy Hall


Buses depart from Centara Grand and Holiday Inn hotels Outdoor Reception Dinner Buses return to Centara Grand and Holiday Inn hotels

Wat Arun, Bangkok Yai

AIDS Vaccine 2011

PROGRAM / TUESDAY

Program
Wednesday, 14 September 2011
08:00 18:30 07:00 17:00 08:30 10:00 08:30 09:00 Registration Speaker Check-In PROGRAM / WEDNESDAY

Wednesday, 14 September

Ballroom Foyer, Level 22 Media Room, Level 22 Ballroom A, Level 22

Plenary Session 02:

New Prevention Strategies


Chairs: Catherine Hankins and Manit Teeratantikanont

Voluntary Medical Male Circumcision for HIV Prevention: PL02.01 What Are the Lessons for Other Prevention Strategies? Helen Weiss London School of Hygiene & Tropical Medicine London, United Kingdom Lessons Learnt from HIV Pre- And Post-Infection Studies: PL02.02 Implications for Combination Prevention Strategies Carolyn Williamson University of Cape Town Cape Town, South Africa Whats New in Preventing Mother-to-Child HIV-1 Transmission (MTCT) PL02.03 Lynne Mofenson National Institute of Child Health and Development, National Institutes of Health Rockville, MD, USA Tea and Coffee Break Ballroom B, Level 22

09:00 09:30

09:30 10:00

10:00 10:30

10:30 12:30

Symposium 01:

Bridging and Sustaining Community Partners in AIDS Vaccine Research


Chairs: Supachai Rerks-Ngarm and Steven Wakefield

Ballroom A, Level 22

10:30 10:54

Combination Prevention Where Clinical Trials and Public Health S01.01 Are at Crossroads A. Cornelius Baker NIAID HIV Vaccine Research Education Initiative/FHI Washington, DC, USA Changing Standards of Care in South Africa: Considerations S01.02 for HIV Vaccine Trials Glenda Gray Perinatal HIV Research Unit Soweto, South Africa Community Engagement for Care in HIV Vaccine Trials: An Exploration S01.03 of Ethical Considerations and Stakeholder Practices/Perspectives Catherine Slack HIV AIDS Vaccines Ethics Group UKZN Pietermaritzburg, South Africa

10:54 11:18

11:18 11:42

AIDS Vaccine 2011

Wednesday, 14 September
11:42 12:06

Program
PROGRAM / WEDNESDAY

In It for the Long Run: Calibrating Community Engagement Efforts to S01.04 Local Contexts and New Challenges Alexandre Menezes International AIDS Vaccine Initiative Rio de Janeiro, Brazil Funding and Sustaining Community Engagement Science S01.05 Partnerships, Politics and Power Steven Wakefield HIV Vaccine Trials Network Seattle, WA, USA

12:06 12:30

10:30 12:30 10:30 10:45

Oral Abstract Session 07:


B Cell Immunology
Chairs: Nicos Karasavva and Bonnie Mathieson

Lotus 1 - 4, Level 22

Bispecific Fusion Antibodies PG9-Ibalizumab (PG9-iMab) and OA07.01 VRC01-Ibalizumab (VRC01-iMab) Exhibit 100% Breadth and Picomolar Potency CS Pace, R Song, D Franco, M Seaman, DD Ho Identification of Amino Acid Residues in HIV-1 Envelope Targeted by OA07.02 Plasma Broadly Neutralizing Antibodies using Evolutionary Models M Lacerda, New Investigator Award Winner PL Moore, N Ngandu, ES Gray, K Wibmer, M Nonyane, D Sheward, BT Korber, DC Montefiori, C Williamson, L Morris, C Seoighe Isolation of a Subset of Novel HIV-1 Broadly Neutralizing Antibodies OA07.03 with Dual Specificity Overlapping the Env Receptor and Coreceptor Binding Sites Y Li, S ODell, X Wu, S Schmidt, C Hogerkorp, Y Feng, B Chakrabarti, R Wilson, N Longo, N Doria-Rose, M Roederer, M Connors, R Wyatt, J Mascola Neutralizing Antibodies Inhibit HIV-1 Transfer from Dendritic OA07.04 Cells to Primary CD4 T Lymphocytes B Su, New Investigator Award Winner K Xu, M Peressin, A Lederle, S Schmidt, G Laumond, C Moog, V Holl Induction of Neutralizing Antibody Responses in HIV Infection Is OA07.05 Associated with HIV-Specific CD4 T Cell Responses M Mueller, M Lindqvist, D Soghoian, S Ranasinghe, T Wrin, P Phung, S Deeks, C Pilcher, F Hecht, H Streeck Influence of NK Cells and NK Cell Receptor Polymorphisms in the OA07.06 Assessment of HIV-1 Neutralization BK Brown, L Wieczorek, G Kijak, K Lombardi, J Currier, M Wesberry, C Ochsenbauer, JC Kappes, V Ngauy, M Marovich, N Michael, DC Montefiori, VR Polonis The Effect of HIV-1 Superinfection on the Development of OA07.07 Neutralizing Antibody Breadth Among Female Sex Workers in Mombasa, Kenya VC Cortez, J Overbaugh The Thai Phase III Clinical Trial (RV144) Induces the OA07.08 LB Generation of Antibodies That Target a Conserved Region Within the V2 Loop of gp120 N Karasavvas, E Billings, M Rao, J Currier, NL Michael, S Rerks-Ngarm, P Pitisuttithum, J Kaewkungwal, V Ngauy, S Nitayaphan, S Madnote, D Arworn, J Kim, M de Souza AIDS Vaccine 2011 9

10:45 11:00

11:00 11:15

11:15 11:30

11:30 11:45

11:45 12:00

12:00 12:15

12:15 12:30

Program
10:30 12:30 PROGRAM / WEDNESDAY 10:30 10:45

Wednesday, 14 September
Lotus 5 - 7, Level 22

Oral Abstract Session 08:

Animal Models and HIV Transmission


Chairs: Stephen Kent and Harriet Robinson

Evidence for CD4 T-Cell Mediated Immune Control of Macrophage OA08.01 Infection in Rhesus Macaques Undergoing CD4+ Lymphocyte Depletion Prior to SIV Infection A Ortiz, B Li, NR Klatt, B Lawson, E Ryzhova, P Carnathan, M Paiardini, J Else, F Gonzalez-Scarano, SJ Ratcliffe, J Brenchley, J Estes, C Derdeyn, G Silvestri CD4+ T Cell Escape in a SIVmac239-Infected Indian Rhesus Macaque OA08.02 Elite Controller with Breakthrough Viremia J Giraldo-Vela, FA Norante, AT Bean, EJ Leon, R Rudersdorf, NA Wilson, DI Watkins, JB Sacha Decreased Penetration of GFP-Labeled Lentiviral Particles in the OA08.03 Vagina and Endocervix of SIV3-Vaccinated Macaques A Carias, M Anderson, E Okocha, M McRaven, W Li, RP Johnson, T Hope Long-Term Programming of Antigen-Specific Immunity from OA08.04 Gene Expression Signatures in the PBMC of Rhesus Macaques Immunized with an SIV DNA Vaccine JD Boyer, SE Belisle, J Yin, DJ Shedlock, A Dai, J Yan, MG Lewis, H Andersen, JA Karl, DH OConnor, A Khan, N Sardesai, RE Palermo, DB Weiner, MG Katze Compromised NK Cell-Mediated Antibody-Dependent Cellular OA08.05 Cytotoxicity in Chronic SIV Infection X He, Z Luo, M Jia, Y Li, Z Cong, D Li, H Liang, Y Shao Cytotoxicity Capacity of SIV-Specific CD8+ T Cells Against Primary OA08.06 Autologous Targets Correlates with Immune Control in SIV-Infected Rhesus Macaques D Mendoza, S Johnson, B Peterson, N Doria-Rose, G Franchini, D Schneider, MT Trivett, CM Trubey, V Coalter, DI Watkins, JD Lifson, SA Migueles, M Connors Live Attenuated Influenza-SIV Vaccines: Efficacy Against Mucosal OA08.07 Challenge in Macaques SJ Kent, S Jegaskanda, J Reece, T Ameresena, A Sexton, C Fernandez, J Stambas, A Brooks, R De Rose Reduction of Founder Virus in Vaccinated Monkeys after Mucosal SIV Challenge JB Whitney, M Rolland, M Lacerda, PT Hraber, A DeCamp, C Luedemann, SS Rao, JR Mascola, B Korber, G Nabel, P Gilbert, C Seoighe, NL Letvin Lunch Buffet Young and early-Career Investigators Networking Lunch OA08.08 LB

10:45 11:00

11:00 11:15

11:15 11:30

11:30 11:45

11:45 12:00

12:00 12:15

12:15 12:30

12:30 13:30

Ballroom B, Level 22

Lunch tables in Ballroom B have been reserved for young and early-career investigators to network with senior researchers. A list of featured researchers is available at the registration desk.

10

AIDS Vaccine 2011

Wednesday, 14 September
13:30 15:00 13:30 13:45

Program
Ballroom A, Level 22 PROGRAM / WEDNESDAY

Oral Abstract Session 09:

Pre-Clinical and Early Clinical Trials


Chairs: Susan Buchbinder and Saladin Osmanov

T-Cell Responses Induced by Immunization with Ad35-HIV-1 OA09.01 Vaccines Are Broad, Durable, Polyfunctional and Can Mediate Inhibition of HIV JH Cox, P Hayes, E Cormier, D Gill, J Kopycinski, A Spentzou, L Hachaambwa, C Bunce, E Sayeed, A Lombardo, W Komaroff, K Loughran, B Barin, L Dally, J Ackland, T Tarragona, M Keefer, J Excler, P Fast, J Gilmour ADCC Titers to HIV-Infected Cells Are Detectable in the Majority OA09.02 of Vaccine Recipients in the RV144 Trial MD Alpert, JD Harvey, WJ Neidermyer, Y Huang, D Morris, L Harris, H Gao, DC Montefiori, P Pitisuttithum, J Kaewkungwal, S Nitayaphan, S Rerks-Ngarm, NL Michael, JH Kim, DT Evans Polyfunctional CD4/CD8 HIV-Specific Cytokine Responses and OA09.03 Presence of Cytolytic Markers in Swedish Vaccinees Immunized with HIV-1 DNA and HIV-1 MVA K Godoy-Ramirez, C Nilsson, K Karln, T Tecleab, M Marovich, B Hejdeman, B Wahren, E Sandstrm, G Biberfeld Surface Plasmon Resonance Analysis of Anti-gp120 V2-Specific OA09.04 IgG Antibodies Generated in the RV144 Thai Trial EA Billings, N Karasavvas, MS de Souza, J Currier, P Pitisuttithum, J Kaewkunwal, S Nitayaphan, PB Gilbert, GD Tomaras, SB Zolla-Pazner, BF Haynes, NL Michael, S Rerks-Ngarm, JH Kim, M Rao V2-Reactive Antibodies in RV144 Vaccinees Plasma OA09.05 S Zolla-Pazner, T Cardozo, A deCamp, B Haynes, J Kim, X Kong, N Michael, S Rerks-Ngarm, C Williams PedVacc001:Safety and Immunogenicity of a Candidate OA09.06 HIV-1 Vaccine, MVA.HIVA, Administered to Healthy Gambian Infants Born to HIV-1/2-Uninfected Mothers MO Afolabi, New Investigator Award Winner J Ndure, J Mueller, V Thomas, M Shams-Rony, N Borthwick, A Bridgeman, A Black, M Reilly, W Jaoko, G John-Stewart, G Ambler, B Lohman-Payne, T Hanke, KL Flanagan

13:45 14:00

14:00 14:15

14:15 14:30

14:30 14:45

14:45 15:00

AIDS Vaccine 2011

11

Program
13:30 15:00 PROGRAM / WEDNESDAY 13:30 13:45

Wednesday, 14 September
Lotus 1 - 4, Level 22

Oral Abstract Session 10:


B Cell Vaccine Concepts
Chairs: Suwat Chariyalertsak and Peter Liljestrom

Eliciting Broadly Neutralizing Antibodies Against HIV-1 Using OA10.01 a Novel Antigen Containing gp41 Membrane-Proximal External Region D Han, H Habte, Y Qin, K Takamoto, C Labranche, D Montefiori, M Cho Neutralizing Antibody Responses Induced with V3-Scaffold Protein OA10.02 Boosts Following Priming with gp120 DNA S Zolla-Pazner, X Kong, X Jiang, T Cardozo, A Nadas, S Cohen, M Totrov, MS Seaman, S Wang, S Lu Phase I Proof of Principle: Immunization with Virosome-gp41-Derived OA10.03 Antigen Induces Mucosal Antibodies with Antiviral Properties G Leroux-Roels, C Maes, F Clement, F van Engelenburg, M van den Dobbelsteen, R Zurbriggen, M Amacker, M Adler, M Spengler, J Detmers, F Anjuere, L Lopalco, D Tudor, A Drillet, M Bomsel, A Chalifour, S Fleury DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge OA10.04 with Heterologous SIVsmE660 R Jalah, V Patel, V Kulkarni, A Valentin, C Alicea, M Rosati, A von Gegerfelt, NY Sardesai, J Bess, Jr., JD Lifson, B Keele, RR Amara, HL Robinson, VM Hirsch, Y Guan, D Venzon, DC Montefiori, BK Felber, GN Pavlakis Direct Antibody Access to the HIV-1 MPER Positively Correlates with OA10.05 Neutralization Sensitivity BK Chakrabarti, LM Walker, JF Guenaga, A Ghobbeh, P Poignard, DR Burton, RT Wyatt A DNA Prime/Protein Boost Vaccine Leads to Higher B-Cell Responses OA10.06 than a Vector Prime/Protein Boost or DNA Prime/Vector Boost Regimens N Frahm, DP Friedrich, P Walsh, S De Rosa, P Spearman, S Barnett, M Keefer, P Goepfert, H Robinson, B Graham, H Liao, G Tomaras, B Haynes, S Rerks-Ngarm, J Kim, MJ McElrath

13:45 14:00

14:00 14:15

14:15 14:30

14:30 14:45

14:45 15:00

13:30 15:00 13:30 13:45

Oral Abstract Session 11:


Mucosal Immunity
Chairs: Aggrey Omu Anzala and Robin Shattock

Lotus 5 - 7, Level 22

Microarray Analysis of Cervical Tissue Reveals a OA11.01 Transcriptional Imprint that Correlates with Protective Immunity in SIV-Nef-Vaccinated Animals AJ Smith, SW Wietgrefe, CS Reilly, L Duan, Q Li, M Zeng, AT Haase Gut Inflammation and Indoleamine Deoxygenase Inhibit IL-17 OA11.02 Production and Induce Cytotoxicity in RORt+ NKp44+ Mucosal NK Cells During SIV Infection R Reeves, P Rajakumar, TI Evans, M Connole, J Gillis, Y Kuzmichev, R Johnson

13:45 14:00

12

AIDS Vaccine 2011

Wednesday, 14 September
14:00 14:15

Program
PROGRAM / WEDNESDAY

Targeting the Vaginal Mucosa with Human Papillomavirus OA11.03 Pseudovirion Vaccines Delivering SIV DNA SN Gordon, RC Kines, G Kutsyna, Ma, A Hryniewicz, BF Keele, JN Roberts, C Fenizia, Hidajat, N Cuburu, B Buck, ML Bernardo, M Robert-Guroff, J Miller, BS Graham, DR Lowy, T Schiller, G Franchini Durability and Phenotype of SIV-Specific Mucosal T Lymphocyte OA11.04 Responses Elicited by Recombinant Adenovirus Vectors in Rhesus Monkeys H Li, J Liu, A Carville, KG Mansfield, S Clark, D Lynch, DH Barouch Role of Genital Tract Inflammation and T Cell Activation in CD4 OA11.05 Depletion at the Female Genital Tract During HIV Infection L Liebenberg, H Jaspan, D Coetzee, A Williamson, J Passmore SIV attenuated vaccine-induced plasma cells and antibodies limit infection at the portal of entry and systemically following vaginal challenge M Zeng, L Duan, Q Li, A Smith, S Wietgrefe, Pj Southern, CS Reilly, S Pambuccian, L Shang, PJ Skinner, M Zupancic, M Piatak, Jr, D Waterman, K Reeves, JD Lifson, P Johnson, AT Haase Poster Viewing with Coffee Break OA11.06 LB

14:15 14:30

14:30 14:45

14:45 15:00

15:00 16:00

Ballroom B, Level 22

16:00 18:00 16:00 16:24

Symposium 02:

Recent Advances in B Cell and Protective Antibody Responses


Chairs: Jos Esparza and Lynn Morris

Ballroom A, Level 22

Mucosal gp41 Antibodies-Mediated Protection from Vaginal S02.01 SHIV Challenges Morgane Bomsel Institut Cochin-Paris Descartes-Inserm U1016 CNRS UMR8104 Paris, France What is a Protective Antibody Response? S02.02 Robert Gallo Institute of Human Virology-University of Maryland School of Medicine Baltimore, MD, USA Efficacy of Non-Neutralizing Antibodies in NHP S02.03 Robin Shattock Imperial College London London, United Kingdom Developing Broadly Neutralizing Antibodies from Between the Gaps S02.04 of the Quantized Humoral Immune Response Peter Kwong National Institutes of Health/NIAID/VRC/SBS Bethesda, MD, USA Broad Neutralization Coverage of HIV by Highly Potent Antibodies S02.05 Pascal Poignard International AIDS Vaccine Initiative (IAVI) New York, NY, USA

16:24 16:48

16:48 17:12

17:12 17:36

17:36 18:00

AIDS Vaccine 2011

13

Program
16:00 18:00 PROGRAM / WEDNESDAY

Wednesday, 14 September
Lotus 1 - 4, Level 22

Symposium 03:

The Role of Host Genetics in Innate and Adaptive Immunity


Chairs: Jacques Fellay and Masafumi Takiguchi

16:00 16:24

The Role of the MHC in Adaptive Immunity Against HIV S03.01 Mina John Institute of Immunology and Infectious Diseases Perth, Australia Host Genetic Factors That Influence HIV-1 Control S03.02 Paul de Bakker Brigham and Womens Hospital Boston, MA, USA Next Generation Sequencing in HIV Host Genetics S03.03 Kevin Shianna Duke University Durham, NC, USA Antiviral Activity of NK Cells in HIV-1 Infection S03.04 Marcus Altfeld Ragon Institute of MGH, MIT and Harvard Charlestown, MA, USA TRIM5 Polymorphism and Experimental Models of HIV Infection and AIDS S03.05 Welkin Johnson Harvard Medical School NEPRC Southborough, MA, USA

16:24 16:48

16:48 17:12

17:12 17:36

17:36 18:00

16:00 18:00 16:00 16:24

Symposium 04:

Integrated and Emerging Novel Issues in Clinical Trials


Chairs: Patricia Fast and Margaret Johnston

Lotus 5 - 7, Level 22

Passive Immunization to Prevent HIV Infection: Potential Applications S04.01 and Challenges Barney Graham Vaccine Research Center, NIH, NIAID Bethesda, MD, USA Development of Ibalizumab and Improved Variants for Passive S04.02 Immunization Against HIV David Ho Aaron Diamond AIDS Research Center New York, NY, USA Challenges in the Design of Future Combination Prevention Trials S04.03 Magdalena Sobieszczyk Columbia University New York, NY, USA

16:24 16:48

16:48 17:12

14

AIDS Vaccine 2011

Wednesday, 14 September
17:12 17:36

Program
PROGRAM / WEDNESDAY

Regulatory Considerations for the Approval of Novel HIV Vaccine S04.04 Clinical Trial Designs in a Developing Country Gavin Steel Management Sciences for Health (MSH) Beacon Bay, South Africa Challenges of Integrating Mucosal Immune Assessments Into S04.05 HIV Vaccine Trials Aggrey Omu Anzala Kenya AIDS Vaccine Initiative / University of Nairobi Nairobi, Kenya

17:36 18:00

18:00 19:30

Poster Session 02 and Reception

Ballroom B, Level 22

All posters ending in even numbers will be presented during Poster Session 02. Presenting authors will be available for discussions at this time. All posters must be removed at the conclusion of Poster Session 02. Remaining posters will be discarded by the convention center staff following the reception.

AIDS Vaccine 2011

15

Program
Thursday, 15 September 2011
08:00 13:30 08:30 10:30 08:30 08:54 Registration

Thursday, 15 September

Ballroom Foyer, Level 22 Ballroom A, Level 22

Symposium 05:
Systems Biology
Chairs: Daniel Douek and Prasert Thongcharoen

Systems Biology of Dengue Infection S05.01 Marcin Kwissa Emory Vaccine Center, Emory University Atlanta, GA, USA Whole Blood Transcriptional Monitoring of Acute HIV-1 Infection S05.02 Reveals Differential Signatures of Host Immune Activation Jason Skinner Baylor Institute for Immunology Research Dallas, TX, USA Functional Genomic Analysis of the Human T Cell Response to Chronic S05.03 Viral Infection William Haining Dana-Farber Cancer Institute Boston, MA, USA Correlates of Immune Protection: A System Biology Analysis of Vaccine S05.04 and Natural Mediated Immune Protection Rafick-Pierre Sekaly Vaccine & Gene Therapy Institute of Florida Port St Lucie, FL, USA Systems Vaccinology: Using the Tools of Systems Biology to Enable S05.05 Rational Vaccine Design Alan Aderem Seattle Biomedical Research Institute Seattle, WA, USA

08:54 09:18 PROGRAM / THURSDAY

09:18 09:42

09:42 10:06

10:06 10:30

08:30 10:30 08:30 08:54

Symposium 06:

New Concepts in Protection Against HIV Acquisitions


Chairs: Amorn Leelarasamee and Georgia Tomaras

Lotus 1 - 4, Level 22

Harnessing Antibody-Mucus Interactions to Prevent HIV Transmission S06.01 Thomas Hope Northwestern University Chicago, IL, USA a47/CD4 T Cells are Key Targets in Mucosal Transmission of HIV S06.02 James Arthos National Institute of Allergy and Infectious Diseases (NIAID) Bethesda, MD, USA

08:54 09:18

16

AIDS Vaccine 2011

Thursday, 15 September
09:18 09:42

Program

Critical Role of Retinoic Acid and TLR Signals in Gut-Associated S06.03 Dendritic Cell Differentiation Jorge Mora Massachusetts General Hospital, Harvard Medical School Boston, MA, USA Defining the Spectrum of Vaccine-Induced Humoral Responses S06.04 Georgia Tomaras Duke University Durham, NC, USA Correlates of Protection Against SIVmac251 in Vaccinated Rhesus Monkeys S06.05 Dan Barouch Beth Israel Deaconess Medical Center Boston, MA, USA

09:42 10:06

10:06 10:30

Characterization of Breakthrough Viruses


Chairs: Jerome Kim and Julie McElrath

08:30 08:54

A Deeper Look At The Virus Populations Infecting Step Trial Volunteers S07.01 James Mullins University of Washington Seattle, WA, USA Sequence Analysis of HIV-1 Breakthrough Infections in the RV144 Trial S07.02 Morgane Rolland U.S. Military HIV Research Program, Henry Jackson Foundation Rockville, MD, USA Increased Multiple Variant Transmission in VAX 003 Injection Drug Users S07.03 Katharine Bar University of Alabama at Birmingham Birmingham, AL, USA Sieve Analysis of RV144 S07.04 Paul Edlefsen Fred Hutchinson Cancer Research Center Seattle, WA, USA Understanding HIV Transmission Utilizing Transmitted/Founder Viruses S07.05 Brandon Keele SAIC-Frederick, National Cancer Institute Frederick, MD, USA Tea and Coffee Break Ballroom B, Level 22

08:54 09:18

09:18 09:42

09:42 10:06

10:06 10:30

10:30 11:00

AIDS Vaccine 2011

17

PROGRAM / THURSDAY

08:30 10:30

Symposium 07:

Lotus 5 - 7, Level 22

Program
11:00 12:30

Thursday, 15 September
Ballroom A, Level 22

Plenary Session 03:

Novel Immunogens and Vaccine Delivery Strategies: Where Do We Go Next?


Chairs: Wayne Koff and Yiming Shao

11:00 11:30

Development of HIV-1 Neutralizing Antibody Vaccines: PL03.01 From Structure to Immunogen Design Jeffrey Boyington Vaccine Research Center, National Institutes of Health Bethesda, MD, USA Current Status and Future Directions of Vector-Based HIV Vaccines PL03.02 C. Richter King International AIDS Vaccine Initiative New York, NY, USA New Generation of Adjuvants for HIV Vaccines PL03.03 Steven Reed Infectious Disease Research Institute Seattle, WA, USA

11:30 12:00

12:00 12:30 PROGRAM / THURSDAY

12:30 13:00 12:30 12:45

Closing Session
Closing Remarks Pratap Singhasivanon 2011 Conference Chair Faculty of Tropical Medicine, Mahidol University Bangkok, Thailand AIDS Vaccine 2012 Dan Barouch 2012 Conference Co-Chair Beth Israel Deaconess Medical Center Boston, MA, USA

Ballroom A, Level 22

12:45 13:00

18

AIDS Vaccine 2011

Special Session SS01.01


Are There Lessons from Past Vaccines for an AIDS Vaccine?
S. Plotkin1
1 University of Pennsylvania and Vaxconsult, Philadelphia, PA, USA

Invited Speakers
SS01.02
Moving Forward from RV144: An Industrial Perspective
S. Gurunathan1, J. Tartaglia1
1

sanofi pasteur, Swiftwater, PA, USA

Developers of AIDS vaccines face unprecedented problems of rapid pathogenesis and wide variation of wild viruses. Thus, it may be presumptuous to think that vaccines for other diseases can teach us something about the future hoped-for prevention of AIDS. Nevertheless, I propose that there are lessons from the importance of mucosal antibody induction by HPV and live influenza vaccines, from the modification of disease even if infection is not prevented in the case of many vaccines, the efficacy of antibodies in vaccination against cytomegalovirus despite the importance of cellular immunity in recovery, the necessity of boosters for vaccines like pertussis and meningococcal, and the problem of attaining high coverage for vaccines given after infancy.

Sanofi pasteur has participated towards the development of an effective HIV vaccine for over two decades. Following the breakthrough results of the phase III HIV vaccine study, RV 144, in Thailand, the study partnership is moving rapidly forward with its commitment.

AIDS Vaccine 2011

19

SPECIAL SESSION

Invited Speakers
Opening Ceremony OC01.01
Progress Towards an HIV Vaccine Lessons Learned from HIV Prevention
C. Dieffenbach1
1

Division of AIDS, NIAID, NIH, Bethesda, MD, USA

As we enter the fourth decade of the HIV/AIDS epidemic it is clear that we have made progress against this disease. In many parts of the world HIV prevention is beginning to have an impact on the growth of the epidemic, but in other nations it seems as if the epidemic is just ramping up. I will review the status of the HIV prevention field and discuss the lessons learned, which inform HIV vaccine discovery and development and highlight the exciting new era of HIV vaccine research and the centrality of vaccines to a global HIV prevention strategy.

OPENING CEREMONY 20 AIDS Vaccine 2011

Invited Speakers
Plenary Session 01: Novel Approaches in Clinical Evaluation through Global Collaboration
PLENARY SESSIONS 21

PL01.01
Africas Contribution to AIDS Vaccine Research and Development
P. Kaleebu1
1

PL01.02
Thailands Contribution to HIV Vaccine Research and Development
P. Pitisuttithum1
1 Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

MRC/UVRI Research Unit on AIDS, Entebbe, Uganda

Background: Sub-Saharan Africa continues to be the hardest hit by the HIV epidemic, contributing 70% of new infections globally in 2010. Existing proven interventions have not sufficiently controlled the epidemic. Additional prevention tools such as vaccines are therefore urgently required. There are scientific reasons to justify African involvement in AIDS vaccine research including differences in HIV subtypes and host genetic composition, pre-existing immunity to potential vectors, existence of high risk cohorts and the need to build local capacity. Achievements: Cohorts were set up in various populations to study HIV trends and to prepare for intervention studies. We have documented in some of the cohorts incidence above 2 per 100 person years and with good retention rates suitable for efficacy trials. These cohorts have provided key information to understand the events around transmission, disease progression and possible protective immune responses. Since the first trial in 1999, approximately 25 phase I and II trials have been conducted in Africa with the first vaccine based on a subtype A strain common in Africa conducted in 2001. A subtype C based vaccine designed in South Africa has also undergone clinical trials. South to South and North to South collaborations have been set up with most funding from the North. Tremendous clinical, statistical, laboratory, regulatory and ethical capacity have been established in many centers in Africa. Challenges: The limited number of promising vaccines is a challenge. There has been reduced funding recently to some institutions involved in HIV vaccine research in Africa and a funding tilt to treatment than prevention. In addition, as circumcision, more widespread and effective antiretroviral treatment and antiretroviral-based prevention technologies become available the incidence of new HIV infections will fall. This will necessitate new strategies or larger efficacy trials, for vaccines to be proven effective.

With the launch of the national AIDS plan by the Ministry of Public Health, and its commitment, especially in relation to disease control and prevention, the reported adult prevalence of HIV in 2009 was 1.3%, and the number of new HIV/AIDS cases had declined to about 10,000 persons per year, in 2010. However, there was an increasing trend among men having sex with men, and younger age groups. The prevalence of HIV among men having sex with men was reportedly 17.3% in 2010. Thailand is the only country to have conducted two efficacy, and many phase I/II, trials involving nearly 20,000 participants. Over the past 18 years, this has become the largest number of participants participating in HIV vaccine research and development anywhere in the world. This has been achieved through national and international collaborations of both private- and public-sector organizations, especially the US Military Research Program and National Institutes of Health. The Thai AIDS Vaccine Evaluation Group (TAVEG) was formed in 1996, by various national academic institutes, including Chiang Mai University, Mahidol University, and the Armed Forces Research Institute of Medical Sciences (AFRIMS) (both Thai-US components). The first efficacy trial conducted by Bangkok Metropolitan Administration, Mahidol University, U.S. Centers for Disease Control and Prevention and Vaxgen Inc. using AIDSVAX B/E alone among injecting drug users, in 1999-2003, showed no efficacy. In 2003, TAVEG and the Ministry of Public Health (MOPH-TAVEG) launched the largest efficacy trial in a low-risk community population. Recently, Chulalongkorn University and the Thai Red Cross Society have joined, to continue HIV/AIDS vaccine research and development efforts. The results of the largest community trial (RV144) using HIV vaccines (ALVAC-HIV vaccine priming and AIDSVAX vaccine boosting) have shown modest (31.2%) protection, for the first time. This has led to tremendous efforts involving scientists and researchers worldwide, to search for antibody correlates of protection, and to improve on the next generations of vaccines.

AIDS Vaccine 2011

Invited Speakers
Plenary Session 01: Novel Approaches in Clinical Evaluation through Global Collaboration PL01.03
Update on RV144: Correlates, Sieve Analysis, and Clinical Development
J. Kim1, MOPH-TAVEG Collaboration1 Walter Reed Army Institute of Research (WRAIR), Rockville, MD, USA
1

PL01.04
Case Control Study of the RV144 Trial for Immune Correlates: The Analysis and Way Forward
B. Haynes1
1

PLENARY SESSIONS

Duke Human Vaccine Institute, Durham, NC, USA

In the two years since RV144 several different groups of collaborators have worked to try to determine a vaccine-induced immune correlate that was associated with decreased risk of infection. These groups have greatly increased our knowledge of the antigenic properties of the vaccine, and started to explore how these antigenic properties induced particular immune responses. Simultaneously, a process for evaluating different laboratory assays for their utility in the correlates analysis yielded a set of primary and secondary (exploratory) analyses that are a part of the formal correlates analysis. In addition, viral sequence analysis is starting to evaluate changes in viral sequences that may correspond to escape or sieving through vaccine-induced defenses. Clinically, a partnership to advance the pox-protein vaccine has been formed and is working to identify the vaccines, populations, and trial designs to advance these vaccines to eventual licensure. Finally, new vaccine concepts aimed at more rapid testing/screening of HIV-1 vaccines and at a global (rather than regional) HIV vaccine form the basis for the next generation vaccine.

The RV144 HIV-1 vaccine efficacy trial showed a modest level of vaccine efficacy. A large collaborative study has been performed that involved investigators from laboratories around the world to determine correlates of protective immunity in the trial. The results of this case controlled study will be presented, and the plans for future studies described.

22

AIDS Vaccine 2011

Invited Speakers
Plenary Session 01: Novel Approaches in Clinical Evaluation through Global Collaboration
PLENARY SESSIONS AIDS Vaccine 2011 23

PL01.05
Poxvirus Vector-Based HIV Vaccines Beyond RV144
G. Pantaleo1
1 Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Switzerland

The field of HIV vaccine has gained re-born enthusiasm from the recent results of the RV144 phase III trial performed in Thailand. The RV144 trial evaluated the efficacy of ALVAC-HIV (vCP1521) in combination with a recombinant gp120 subunit vaccine (AIDSVAX B/E). The results indicated that this vaccine combination was effective in preventing infection, i.e. 31.2% efficacy, while it showed no effect on the levels of viremia and/or CD4 T-cell count during the acute phase of infection in vaccinated subjects who were subsequently diagnosed with HIV-1. These results, although showing a modest efficacy, are important since they demonstrate for the first time that an HIV vaccine is capable of preventing HIV infection. A clear message from the RV144 trial is that both components of the vaccine, i.e. the priming component (ALVAC) and the boosting component (the gp120 protein), need to be substantially improved. The improved vaccine combinations should aim at eliciting an integrated immune response, i.e. innate plus humoral plus cellular immunity. Replication deficient adenovirus and poxvirus vectors and plasmid DNA have been predominantly used as priming component for protein boost or in prime boost combinations and have represented the lead strategy for the development of T-cell vaccine regimens. Although these vaccines have shown to induce vigorous T-cell responses, they are not optimal priming for protein boost and development of antibody response and there are still some questions open regarding their efficiency in stimulating innate immunity and broad T-cell responses. Recent developments in poxvirus vectors are showing that highly attenuated replication competent virus vectors (NYVAC-KC) induce optimal priming for inducing vaccine integrated immune response, i.e. innate plus cellular plus humoral responses. In particular, substantial improvements have been observed in the ability to efficiently prime for protein boost and for the development of antibody responses. The last 20 years of HIV vaccine research have been predominantly focused to develop virus vector-based vaccines aimed to inducing potent T-cell responses. Future development of virus vectorbased vaccines should be tailored to the stimulation of potent antibody responses. Highly attenuated replication competent poxvirus vectors will play an important role in the development of future immunization regimens aiming to inducing potent antibody response.

Invited Speakers
PL02.01
PLENARY SESSIONS

Plenary Session 02: New Prevention Strategies PL02.02


Lesson Learned from HIV Pre- and Post Infection Studies: Implications for Combination Prevention Strategies
C. Williamson1, CAPRISA Study Team2, and the NIH/NIAID HIV Vaccine Trial Collaborative Network3 Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, South Africa; 2 University of Kwa-Zulu Natal, South Africa; 2National Institute for Communicable Diseases, South Africa; 3 University of Washington, Seattle; Fred Hutchinson Cancer Research Center, Seattle; 3University of Witwatersrand, South Africa
1,2,3

Voluntary Medical Male Circumcision for HIV Prevention: What Are the Lessons for Other Prevention Strategies?
H. Weiss1, C. Hankins2 London School of Hygiene & Tropical Medicine, London, United Kingdom (Great Britain); 2UNAIDS, Geneva, Switzerland
1

Three randomized controlled trials conducted in sub-Saharan Africa have shown that medical male circumcision (MMC) is safe, and can reduce the risk of HIV acquisition in heterosexual men by around 60%. In 2007, the World Health Organization and UNAIDS recommended that male circumcision be included in a comprehensive package of HIV prevention, and voluntary MMC is now being scaled up in several countries in southern and eastern Africa. MMC was the first HIV prevention strategy to be shown to be efficacious in multiple randomized controlled trials. The experience gained from translating the scientific evidence on MMC into public health action will benefit other HIV prevention strategies, including HIV vaccines, pre-exposure prophylaxis, vaginal microbicides. Scaling-up of voluntary MMC has confronted issues common to other biomedical strategies, including safety, acceptability, integration with HIV counseling and testing, partial protection and risk compensation. Additional concerns include the impact on women, cultural sensitivities, dialogue with traditional circumcision providers, the logistics of providing a surgical intervention in under-resourced healthcare systems, and managing the development of new devices to simplify the procedure. Conversely, as a one-time procedure, there are no issues of user-adherence, drug resistance, no need for repeat HIV testing, and MMC is cost-saving.Almost five years after MMC was recommended for HIV prevention, over half a million men in southern and eastern Africa have been circumcised for HIV prevention, and several million males are likely to be circumcised in the next five years, as voluntary MMC is now included in national strategic plans for HIV prevention. In this talk we discuss the lessons learnt to date from scaling-up of circumcision, and the implications for other HIV prevention strategies.

Understanding pre-infection factors that lower the barriers to HIV infection, together with post-infection factors that potentially accelerate or ameliorate disease progression, is crucial to informing prevention strategies. This presentation will overview results from large microbicide and vaccine clinical trials, together with pathogenesis studies, to provide valuable information on the dynamic interplay of immunological damage, transmission, viral evolution and host adaptive immunity. Inflammatory responses prior to infection were found to facilitate transmission thereby lowering the barrier of infection and reducing the effectiveness of the intervention. Where infection occurred, there was no discernable effect on the number of viral variants (genetic bottleneck) associated with transmission in participants from either the 1% Tenofovir CAPRISA 004 microbicide trial or the HVTN503 Phambili vaccine trial. Evidence for a vaccine sieve effect was found in the Phambili trial which tested the MRKAd5 B-clade vaccine in a C-clade epidemic, although this effect was much weaker than previously reported from the STEP trial (Rolland et al., 2011). The potential to enhance protection through modulating immune responses, while eliciting specific cellular and neutralizing antibody responses post-infection that may ameliorate disease progression, will be discussed.

24

AIDS Vaccine 2011

Plenary Session 02:New Prevention Strategies PL02.03


Whats New in Preventing Mother-to-Child HIV-1 Transmission (MTCT)
L. Mofenson1
1 National Institute of Child Health and Development, National Institutes of Health, Rockville, MD, USA

Invited Speakers
PLENARY SESSIONS AIDS Vaccine 2011 25

Globally, 1,000 children are newly infected with HIV-1 daily, most in sub-Saharan Africa; the primary mode of HIV-1 acquisition is through mother-to-child transmission (MTCT) during pregnancy, childbirth or breastfeeding. In well-resourced health systems like the United States, universal HIV-1 testing of pregnant women, provision of antiretroviral (ARV) therapy (when needed for maternal health) or prophylaxis, elective cesarean delivery, and avoidance of breastfeeding has reduced MTCT to ~1%. However, in resource-limited countries, the perinatal epidemic continues. Clinical trials have identified simple, less expensive, effective ARV prophylaxis regimens capable of being implemented in resourcelimited settings, but implementation has been slow and breast milk HIV transmission remains a challenge. Randomized trials have demonstrated ARV prophylaxis of the breastfeeding infant or the lactating mother can significantly reduce postnatal HIV-1 transmission while the prophylaxis is given, although infection risk resumes when prophylaxis is stopped. The World Health Organization (WHO) recommends that HIV-infected pregnant women with CD4 350 cells/uL or WHO Stage 3/4 disease start on life-long ARV therapy; ARV prophylaxis should be provided to all HIV-infected pregnant women who do not require therapy starting as early as 14 weeks gestation. WHO recommends countries choose either infant ARV prophylaxis or maternal triple ARV prophylaxis during 12 months of breastfeeding. However, in resource-limited countries, breastfeeding is a cornerstone of infant survival, and data suggest that shortened breastfeeding by HIV-1-infected woman, while reducing HIV transmission, results in decreased infant survival. Thus, there is a critical need for interventions to allow safer and more prolonged breastfeeding into the second year of life. Even with combined use of antepartum and postnatal ARV prophylaxis, overall MTCT rates are 3-5% at age 12 months. A combined approach of active and passive immune strategies in addition to current ARV interventions could enable prolonged breastfeeding without the potential toxicity of prolonged antiretroviral drug administration.

Invited Speakers
Plenary Session 03: Novel Immunogens and Vaccine Deliver Strategies: Where Do We Go Next? PL03.01
Development of HIV-1 Neutralizing Antibody Vaccines: From Structure to Immunogen Design
J. Boyington1, Z.Y. Yang1, L. Wu1, L. Xu1, W. Akahata1, T. Zhou1, S. ODell1, S. Schmidt1, M.K. Louder1, J.R. Mascola1, P.D Kwong1, G.J. Nabel1
1 Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA

PL03.02
Current Status and Future Directions of VectorBased HIV Vaccines
C. R. King1
1

PLENARY SESSIONS

International AIDS Vaccine Initiative, New York, NY, USA

Advances in our basic scientific understanding of the immune system and mechanisms of HIV pathogenesis have provided the tools to rationally design AIDS vaccine candidates. We have used knowledge of HIV-1 Envelope (Env) structure to develop antigenically resurfaced glycoproteins specific for the structurally conserved site of CD4 receptor binding. These probes identified sera with broadly neutralizing antibodies from infected donors and enabled the isolation of VRC01, a prototypic CD4-binding site (CD4bs) antibody that neutralized over 90% of circulating HIV-1 isolates. The crystal structure of the VRC01 in complex with HIV-1 gp120 core protein was determined and has been used to develop immunogens that re-elicit these antibodies. Candidate immunogens have been generated from rationally designed trimer, resurfaced core, monomer, or outer domain proteins. Specific Env domains have also been presented on highly arrayed alphavirus like particles. Immunization of rodents and NHP has been performed, and CD4bs antibodies of limited breadth have been elicited. Further refinement of this approach has been informed by analysis of the VRC01 related antibody repertoire in HIV-infected subjects.

The use of vectors to deliver immunogens has long been recognized as having many theoretical advantages for the generation of an effective HIV vaccine. These include: the delivery of immunogens in a context similar to a natural virus infection; the delivery of native protein structures or selected epitopes to specific cells or tissues; as well as practical advantages for scale up and quality control. As observed with other vaccine approaches, the development of vectored HIV vaccines has been hampered by the genetic diversity as well as the immune evasion qualities of HIV. Design of vectored vaccines has also been limited by the lack of clearly defined links between elicited immune responses and efficacy. However, recent progress in many areas of HIV vaccine research is providing new insights for design of improved vectored vaccines. Building on the partial protection seen in the RV-144 trial, new clues concerning mechanisms of protection are expected to be identified. Over the next few years, data from further clinical trials of vector-based vaccines can be mined to identify the preferred characteristics of a vectored vaccine. In the preclinical realm, recent discoveries have shown that different vaccine strategies can reduce set point viral load by as much as 4 orders of magnitude. As the analyses of correlates of efficacy proceed, design of improved vectored vaccines is occurring on multiple fronts. Currently available vectored vaccines based on DNA, Adenovirus, and Pox virus vectors are being tested in various prime boost combinations. Delivery of vectors to mucosal surfaces and in combination with adjuvants is also being investigated. The underlying technology for vector delivery of immunogens continues to be enhanced by improvements to existing platforms, as well as the development of novel technologies to incorporate replication competency based on Adeno, Pox, Sendai, Vesicular Stomatitis and Canine Distemper Viruses. Equally important, there is a need for the design and testing of new genetic inserts for these vector platforms. Inserts designed to improve T cell-based immune responses are advancing in clinical trials and new strategies are being proposed and tested. Although design of genetic inserts to elicit effective antibody responses is less advanced, discoveries in the areas of human broadly neutralizing antibodies are expected to provide opportunities to design vectors that deliver immunogens that more closely mimic the HIV envelope. By combining the insights derived from these preclinical and clinical investigations, a new generation of more efficacious vector vaccines for HIV will likely emerge.

26

AIDS Vaccine 2011

Invited Speakers
Plenary Session 03: Novel Immunogens and Vaccine Deliver Strategies: Where Do We Go Next?
PLENARY SESSIONS AIDS Vaccine 2011 27

PL03.03
New Generation of Adjuvants for HIV Vaccines
S. Reed1
1

Infectious Disease Research Institute, Seattle, WA, USA

Safe and effective adjuvants for prophylactic and therapeutic vaccine use are resulting from the identification and optimizing formulations of small molecules. Effectively engaging macrophages and dendritic cells (DC), leading to T cell responses is essential for developing a new generation of T cell vaccines (e.g. tuberculosis, malaria, HIV), as well as for improving the quality and duration of antibody responses (influenza, HPV, HIV, etc.). The most advanced approaches to new adjuvant development consist of using TLR ligands (TLRL), to provide synergism between the formulation and the TLRL. Using the crystal structure of the MD2 molecule of human TLR4, we have designed and optimized a new generation of ligands. Engagement of TLR4 may lead to activation of signaling pathways, or to inhibition of signaling pathways, depending in part on the molecular structure of the TLR ligand, and its interaction with MD2. Position, number, and length of acyl chains present in the TLR4L all influence responses by antigen-presenting cells (APC). Furthermore, varying these factors, e.g. acyl chain number may produce molecules active in mouse, but not human, APC., We have characterized molecular signatures of human dendritic cell (DC) responses using novel TLR4L, alone or in combination, to design adjuvant potency and immune response quality.The manner in which TLRL are formulated dramatically influences the nature of the immune response induced. We have developed formulations of our lead TLR4L, GLA, and have evaluated a variety of these, including oil/ water emulsions, micellar, niosomal, and liposomal, in clinical trials and in a variety of preclinical models. When properly formulated, GLA, which was optimized for activity on human cells, but is also active in animal models, has the ability to focus the T cell to type I, to down regulate type 2 responses, induce CD8 T cells, and effectively enhance and broadened specific antibody responses, including mucosal immune responses. We have also shown that cells from elderly individuals strongly recognize GLA, producing cytokines in a manner qualitatively and quantitatively indistinguishable from cells from healthy adults. Thus, it appears that selective molecular synthesis and formulation may lead to a new generation of TLR4L-based adjuvants with improved qualities over natural products.

Invited Speakers
S01.01

Symposium 01: Bridging and Sustaining Community Partners in AIDS Vaccine Research S01.02
Changing Standards of Care in South Africa: Considerations for HIV Vaccine Trials
G. Gray1
1

Combination Prevention Where Clinical Trials and Public Health Are at Crossroads
A. C. Baker1
1 NIAID HIV Vaccine Research Education Initiative/FHI, Washington, DC, USA

Perinatal HIV Research Unit, Soweto, South Africa

For much of the past 30 years, community leadership has been an essential component of the worldwide response to the AIDS epidemic. Three decades of prevention education have had a demonstrable impact on raising awareness regarding HIV/ AIDS among most populations. However, certain communities still contract HIV in alarming rates. In the United States, this is especially true of African Americans, Latinos/as, and gay men and transgender women of all races. While prevention research has had several milestone advances in recent years, the populations most impacted are still not equitably represented in critical biomedical research trials in the US. Research conducted for the NIAID HIV Vaccine Research Education Initiative (NHVREI) and by others continues to indicate that myths and misinformation about HIV and HIV prevention research persist among populations affected by HIV and AIDS. For example:A household survey conducted in 2003 found that 47 percent of African-American participants as well as 27 percent of Latinos believed that a preventive HIV vaccine already exists. In 2005 other researchers noted the belief among some populations that while a vaccine exists it is being withheld in order to hurt or weaken vulnerable populations. In 2009 focus groups conducted with African Americans, Hispanic/ Latinos, MSM, and transgender individuals in four U.S. cites revealed that almost all individuals had no prior knowledge of HIV vaccine research.As we approach a crossroads in vaccine and other HIV prevention research, these findings raise critical questions: Will a vaccine work for populations most impacted by HIV? What is needed to mobilize the most affected populations to support and/or participate in HIV vaccine research? How will new prevention technologies be used most effectively in combination?The presentation will address these questions and efforts to educate, engage and mobilize the community to ensure the success of future research.

The design of future HIV vaccine efficacy trials will have to incorporate new HIV prevention modalities like microbicides, pre-exposure antiretroviral prophylaxis, male circumcision and antiretroviral interventions that have been found to be effective for HIV prevention in discordant couples. These changing standards of HIV prevention will impact not only on clinical trial designs, but also to their cost, complexity and size. Interventions found to be efficacious may not yet be licensed for use or be available to the general population, but there may be an ethical imperative to make these interventions available to study populations. This presentation will explore the operational challenges, cost, public health issues and regulatory issues that may arise when new biomedical HIV prevention modalities are integrated into HIV vaccine efficacy trials. As new modalities are shown to be efficacious, their limited availability in country due to licensure, production or cost constraints may cause tensions in study populations that will require mitigation, if they are only available to study participants in HIV prevention trials. Where HIV prevention modalities are not widely available in communities, negotiation with government, regulators and community will be paramount to ensure the successful execution of these trials.

SYMPOSIA

28

AIDS Vaccine 2011

Symposium 01: Bridging and Sustaining Community Partners in AIDS Vaccine Research S01.03
Community Engagement for Care in HIV Vaccine Trials: An Exploration of Ethical Considerations and Stakeholder Practices/Perspectives
C. Slack1
1 HIV AIDS Vaccines Ethics Group UKZN, Pietermaritzburg, South Africa

Invited Speakers

S01.04
In It for the Long Run: Calibrating Community Engagement Efforts to Local Contexts and New Challenges
A. Menezes1
1

International AIDS Vaccine Initiative, Rio de Janeiro, Brazil

The HIV/AIDS Vaccines Ethics Group (HAVEG) has been funded by the Wellcome Trust Biomedical Ethics Program to explore care/ prevention responsibilities in HIV vaccine trials in South Africa. This research comprised a conceptual review of normative guidance regarding stakeholders responsibilities to ensure ancillary care in HIV vaccine trials. It comprised an empirical investigation that was exploratory, descriptive, and applied. The empirical research utilized document review and stakeholder interviews to identify reported ancillary care practices, and perspectives, of research stakeholders. It assessed the degree to which stakeholder practices correspond with normative guidance, and stakeholder dilemmas were anticipated in normative guidance. This paper reports on stakeholder practices (and perspectives) regarding community engagement for care issues in such trials, including site staff, CAB members and REC members. Ethical guidelines for care include recommendations that researchers understand care options, and collaborate with available serviceproviders to meet participants care needs; and that community representatives be involved in care decisions, with recent requirements for stakeholder consensus and documentation of discussions (GPP, 2007; UNAIDS, 2007). While protocols and supporting documents do not contain much detail on community engagement activities, sponsor-investigators report numerous steps to partner with care facilities to address care needs (in addition to direct provision of some care); as well as efforts to engage community representatives in protocol development and review. A number of challenges are reported. Interviewees do not report steps to ensure all research stakeholders have reached consensus and report skepticism about the ethical value and feasibility of this requirement. Recommendations for future guideline development are made, including the need to clarify how the normative standard of optimal care should interact with the procedural recommendations to seek consensus on care.

In diverse settings around the world AIDS vaccine research has opened the door for structured dialogue between researchers and the communities that will ultimately benefit from the discovery of a vaccine. This presentation will focus on the different ways by which the community sector has affected the research process in developing countries by contributing to recruitment strategies, working with researchers through consultative mechanisms, helping disseminate accurate information among stakeholders or engaging in independent advocacy. The presentation will examine best practices and current examples of how international models were adapted to developing countries hosting research, ensuring the appropriateness of community engagement efforts to local contexts. In recent years, the continued evolution of these models has been marked by changes in the communities themselves as well as significant transformations in the HIV prevention research field and the priorities for the development of AIDS vaccines. Lessons learned from these processes will be considered to inform a discussion of current needs and future directions in sustaining continued collaboration between researchers and the community sector.

AIDS Vaccine 2011

29

SYMPOSIA

Invited Speakers
S01.05

Symposium 01: Bridging and Sustaining Community Partners in AIDS Vaccine Research

Funding and Sustaining Community Engagement Science Partnerships, Politics and Power
S. Wakefield1
1

HIV Vaccine Trials Network, Seattle, WA, USA

The expansion of HIV vaccine clinical research worldwide requires strong, long-term relationships with communities. The current infrastructure for the engagement of communities involved in HIV vaccine clinical research trials is based in needs for and limited to resources for the recruitment and support of study participants. This talk will look at the need for dedicated resources to support the science of community engagement. Can we ensure consistent, high-quality community engagement efforts and the effective implementation of existing good participatory practice guidelines? Considerations will include formative research activities, stakeholder advisory mechanisms, stakeholder engagement plan, stakeholder education plan, communications plan, issues management plan, site selection, role of stakeholders in protocol development, informed consent process evaluation, responsiveness to changing standards of HIV prevention, access to HIV care and treatment, non-HIV-related care, policies on research-related harms, study accrual, follow-up and exit, trial closure and results dissemination, and future access to new HIV prevention options.

SYMPOSIA

30

AIDS Vaccine 2011

Symposium 02: Recent Advances in B Cell and Protective Antibody Responses S02.01
Mucosal gp41 Antibodies-Mediated Protection from Vaginal SHIV Challenges
M. Bomsel , S. Fleury , F. van Engelenburg , D. Tudor
1 2 3 1 1

Invited Speakers

S02.02
What Is a Protective Antibody Response?
R. Gallo1, G. Lewis1, A. DeVico1
1 Institute of Human Virology-University of Maryland School of Medicine, Baltimore, MD, USA

Institut Cochin-Paris Descartes-Inserm U1016 CNRS UMR8104, Paris, France; 2Mymetcis, Lausanne, Switzerland; 3Kinesis Pharma B.V., Breda, The Netherlands
AIDS is mainly a sexually transmitted disease and mucosal tissues are the primary sites of natural HIV-1 transmission. Mucosal IgA antibody specific for HIV-1 envelope gp41 is one correlate of protection in individuals highly sexually exposed to HIV-1 but remaining persistently IgG seronegative (HEPS). We have build a mucosal Fab IgA library from HEPS and have characterized a series of IgAs specific for gp41 that in vitro are transcytosis-blocking and infection-neutralizing activities. These IgA genes exhibit affinity maturation due to an antigen driven process presumably resulting from multiple exposures to HIV-1. Such knowledge on protective epitopes and antibody-mediated mechanisms of protection was used to design a protective mucosal HIV-1 vaccine prior to establishment of infection and viral reservoirs. Our vaccine is based on conserved gp41-X4 derived antigens critical in HIV-1 entry at mucosal site and infection. These antigens are grafted on virosomes as safe market-approved delivery carrier with self-adjuvant properties. Female Macaca mulatta were immunized by the combined intra-muscular and intra-nasal route to favor a vaginal immune response. The animals were challenged 13 repeated times with a restricted dose of SHIV SF162P3 by the vaginal route. Four out of 5 animals remained virus negative for up to 6 months post challenge, while the 5th animal was only transiently infected. None of the 5 animals seroconverted to p27gag SIV. In contrast, 6 out of 6 placebo-vaccinated animals became infected and seroconverted. All the protected animals showed vaginal gp41-specific IgAs with HIV-1 transcytosis-blocking properties and vaginal IgGs with neutralizing activity and/or antibody-dependent cellular cytotoxicity (ADCC). In contrast, circulating IgGs in the plasma totally lacked virus-neutralizing or ADCC activities. Although actual correlates of protection remain to be firmly established, the protection observed challenges the paradigm whereby circulating neutralizing antibodies are required for protection against HIV-1 infection. A phase I study was conducted with the MPER gp41 subunit-virosome. It opens new strategies for the development of a human vaccine against HIV-1/AIDS.

In AIDS, two major practical achievements have been made: (1) blood test and (2) therapy. Three great needs remain: (1) drug delivery to developing nations (although this has been greatly aided by the U.S. PEPFAR program); (2) a cure so as no further therapy is needed by complete viral eradication or in the absence of a cure the continued development of new approaches to therapy because therapy is life-long and consequently can lead to side effects and HIV drug-resistant mutants; and (3) a successful vaccine. I will specifically describe the difficulties in HIV vaccine development but also some important recent progress. To summarize: the special challenges for a successful HIV vaccine are due to HIV DNA integration, HIV variation, and its early harm to the immune system. Though easy to describe, the challenge is uniquely difficult compared to past successful vaccines. However, most of the current and past vaccine candidates have not taken these features of HIV into account. One exception was the Thai trial conducted by the U.S. Army vaccine program leaders, headed by Dr. Nelson Michael with Thai collaborators and Aventis Sanofi Inc. What is needed and has been needed for over two decades are: (1) far more availability of primates and to a broader number of scientists; (2) an immune response which is sustained; (3) an immune response which is broad and results in sterilizing immunity or close to sterilizing immunity, which clearly must involve the envelope protein of HIV. Finally, I will describe the characteristics, progress, and remaining problems with a candidate vaccine developed at the Institute of Human Virology and what aspects of the ENV used in the Thai trial appear to be similar to our ENV vaccine approach.

AIDS Vaccine 2011

31

SYMPOSIA

Invited Speakers
S02.03

Symposium 02: Recent Advances in B Cell and Protective Antibody Responses S02.04
Developing Broadly Neutralizing Antibodies from Between the Gaps of the Quantized Humoral Immune Response
P. Kwong1
1 National Institutes of Health/NIAID/VRC/SBS, Bethesda, MD, USA

Efficacy of Non-Neutralizing Antibodies in NHP


R. Shattock1
1 Imperial College London, London, United Kingdom (Great Britain)

The field of HIV vaccine research has recently been reinvigorated by the perceived success of the RV144 Thai trial of ALVAC gp120 B/E gp120 protein. Although the level of observed protection was low (31%), it is credited as providing the first potential indication that the development of an effective vaccine might be possible. The prevention of acquisition coupled with the short duration of responses and dearth of CD8 T-cell responses has prompted the hypothesis that protection in RV144 was either due to a short-lived antibody response, an ALVAC-induced short-lived innate response, or a combination of both. The lack of evidence for neutralization as a protective effect in the Thai trial has led to speculation that non-neutralizing gp120 binding antibodies may have contributed to the very modest protection, perhaps by inhibiting or impeding HIV transmission at mucosal surfaces. This in turn has stimulated a resurgence of interest in non-neutralizing antibody (NNAb) in preventing HIV infection. A number of different NNAb functions have been attributed with potential for impacting on transmission events including: ADCC/ ADCVI, inhibition of transcytosis, viral aggregation, mucus trapping and exclusion of virions, Fc-mediated inhibition of viral infection of macrophages and dendritic cells, and complement opsonization. The potential contribution of these mechanisms alone or in concert to reducing transmission is unclear. Furthermore NNAb are not without a dark side, with suggested potential for mediating antibody mediated enhancement of infection, although the in vivo relevance has yet to be defined. NNAb target both functional and non-functional envelope epitopes providing a great breadth of reactivity and potential for targeting conserved epitopes with different density on the virion surface. This presentation will review the latest developments in understanding the role of NNAb in HIV transmission as it relates to functional activity and the potential correlation to protective efficacy in NHP vaccine and passive infusion studies.

The humoral immune system is capable of generating broadly neutralizing antibodies that effectively neutralize HIV-1. Such antibodies, however, have only been identified in infected individuals, are only observed after several years of infection, and appear to require extraordinary degrees of affinity maturation. For example, with the VRC01-like antibodies that target the site of CD4 attachment on HIV-1 gp120, over 100 nucleotide changes occur in the variable regions of these antibodies while progressing from nascent recombinant to potent neutralizer. In 10 ml of blood, there are typically 1 million B cells, and 454 pyrosequencing allows for the sequences of these to be determined. Deep sequencing of donors with broadly neutralizing antibodies allows for the sequences of thousands of broadly neutralizing antibodies to be identified. Analysis of these antibodies for lineage signatures, for example in the 3rd complementary determining regions of the heavy chain (CDR H3) or the light chain (CDR L3), provide genetic roadmaps for the development of these antibodies. These roadmaps can be used in a number of ways: to determine the sequences of putative ancestors, to map the development of broadly neutralizing activity, to infer maturation intermediates, and to trace maturation lineages. Each of the 454 analyses provides a genetic snapshot of the antibodyome. When snapshots are combined with longitudinal sampling of both virus and developing humoral immune system we may have sufficient information to map the antigen-driven interactions that allow for broadly neutralizing antibodies to develop. Through a combination of structural biology and functional genomics, we hope to gain sufficient information to understand how broadly neutralizing antibodies develop and how this development might be stimulated with appropriate immunization.

SYMPOSIA

32

AIDS Vaccine 2011

Symposium 02: Recent Advances in B Cell and Protective Antibody Responses S02.05
Broad Neutralization Coverage of HIV by Highly Potent Antibodies
L.M. Walker1, M. Huber1,7, K. J. Doores1,7, E. Falkowska1,7, R. Pejchal2, J.P. Julien2, S.K. Wang6, A. Ramos1, P.Y. Chan-Hui3, M. Moyle3, J. L. Mitcham3, P.W. Hammond3, O.A. Olsen3, P. Phung4, S. Fling5, C.H. Wong6, S. Phogat5,T. Wrin4, M. D. Simek5, Protocol G Principal Investigators, W. C. Koff5, I. A. Wilson2, D.R. Burton1,7, P. Poignard1,5
1 Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; 2Department of Molecular Biology, Skaggs Institute for Chemical Biology, and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; 3Theraclone Sciences, Inc., Seattle, WA, USA; 4Monogram Biosciences, Inc., South San Francisco, CA, USA; 5International AIDS Vaccine Initiative, New York, NY, USA; 6Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA; 7Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA

Invited Speakers

About 10-30% of HIV-infected individuals develop broadly neutralizing sera, and protective broadly neutralizing monoclonal antibodies (bnMAbs) have been isolated from infected donors. We have previously screened sera from approximately 1,800 HIV infected donors for neutralization breadth and potency, designating the top 1% as elite neutralizers, based on a score incorporating both breadth and potency. We have now probed the neutralizing antibody repertoires of the top four elite neutralizers and rescued 17 new MAbs that neutralize broadly across clades. Many of the new MAbs are almost 10-fold more potent than the recently described PG9, PG16, and VRC01 bnMAbs and 100-fold more potent than original prototype HIV bnMAbs. The MAbs largely recapitulate the neutralization breadth found in the corresponding donor serum and many recognize novel epitopes on the envelope glycoprotein gp120, illuminating new targets for vaccine design. Analysis of neutralization by the full complement of anti-HIV bnMAbs now available reveals that certain combinations of antibodies provide significantly more favorable coverage of the enormous diversity of global circulating viruses than others. In summary, an effective vaccine against HIV will likely require the elicitation of a combination of complementary potent neutralizing antibodies. The demonstration that large numbers of diverse and highly potent bnMAbs can be isolated from several different individuals provides grounds for renewed optimism that an antibody-based vaccine may be achievable.

AIDS Vaccine 2011

33

SYMPOSIA

Invited Speakers
S03.01

Symposium 03: The Role of Host Genetics in Innate and Adaptive Immunity S03.02
Host Genetic Factors That Influence HIV-1 Control
P. de Bakker1
1

The Role of the MHC in Adaptive Immunity Against HIV


M. John
1 1

Brigham and Womens Hospital, Boston, MA, USA

Institute of Immunology and Infectious Diseases, Perth, Australia


A number of genome-wide association studies based on different outcome phenotypes and different population characteristics have shown a remarkable concordance in finding the strongest host determinants of HIV control to be within the major histocompatibility complex, and have driven more recent research on the biological mechanisms underlying the genetic associations. While the protective effects of particular HLA-B alleles are in large part mediated by their distinct interactions with HIV peptide epitopes and HIV-specific CD8 T cells, the allele-specific effects of HLA-C may derive from differential posttranscriptional regulation of HLA-C expression. The challenge for translating these findings to vaccine design is to recapitulate highly host-specific effects found in nature to a genetically diverse population of vaccinees. In addition, the diversity of circulating HIV viruses is influenced by HLA diversity and may even lead to regions of HIV which have adapted to HLA at the population level. Here an understanding of MHC-HLA over a broader evolutionary period may be useful, as like HIV, the HLA system has evolved through a primate lineage. Mapping of regions in the HIV proteome where there is concordance or discordance in conservation across the primate lentivirus phylogeny versus contemporary HIV strains may help to identify HIV immunogen epitopes which are most likely to elicit good T cell responses in the context of a broad range of HLA types and be most robust to HLA-driven adaptation among circulating viruses.

Although it has been very difficult to arrive at accurate heritability estimates of HIV-related phenotypes, it is evident that the HIV exploits various host factors in infection. Host genetic association studies have the power to reveal important host factors that play critical roles in HIV pathogenesis. In this talk, Dr. De Bakker will give an overview of recent genome-wide studies of HIV disease, and explain how other technologies may impact HIV/AIDS research.

SYMPOSIA

34

AIDS Vaccine 2011

Symposium 03: The Role of Host Genetics in Innate and Adaptive Immunity S03.03
Next Generation Sequencing in HIV Host Genetics
K. Shianna1
1

Invited Speakers

S03.04
Antiviral Activity of NK Cells in HIV-1 Infection
M. Altfeld1, G. Alter2, L. Fadda2, D. Heckerman3
1 Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2Ragon Institute, Boston, MA, USA; 3Microsoft Research, Seattle, WA, USA

Duke University, Durham, NC, USA

AIDS Vaccine 2011

35

SYMPOSIA

Genomics research has already made an impressive mark in the HIV field. Recent improvements in next generation sequencing now make it possible to quickly and cost effectively sequence hundreds of human genomes, further entrenching genomics in HIV research. Discovery genetics can now move beyond the study of common variation (GWAS) and can focus on the evaluation of both common and rare variants associated with the many HIV phenotypes that are relevant to the development of an HIV vaccine (vaccine response, viral control, protection, etc). An overview of the current state of next generation sequencing will be provided along with a brief introduction on future sequencing technologies. In addition, two relevant ongoing sequencing studies will be discussed that focus on the identification of genetic variants that are responsible for 1) HIV resistance in highly exposed individuals and 2) rapid HIV progression.

NK cells have been shown to play an important role in the control of viral infections, and increasing data indicate that NK cells might also contribute to the control of HIV-1 infection. Specific NK cell receptor (KIR) genotypes in conjunction with their HLA class I ligands have been associated with slower HIV-1 disease progression, and NK cell subpopulations are expanded in acute HIV-1 infection. Here we demonstrate that HIV-1 is adapting to KIR genotypes at the population level by selecting for sequence polymorphisms that are associated with specific KIRs. In functional studies we furthermore show that these KIR-associated sequence polymorphisms allow for the escape from NK cellmediated recognition of infected cells in vitro. These data suggest that NK cells can mediate antiviral activity against HIV-1, and might contribute to HIV-1 sequence evolution.

Invited Speakers
S03.05

Symposium 03: The Role of Host Genetics in Innate and Adaptive Immunity

TRIM5 Polymorphism and Experimental Models of HIV Infection and AIDS


W. Johnson1
1

Harvard Medical School NEPRC, Southborough, MA, USA

The host antiviral restriction factor Trim5 displays significant interspecies divergence among primates, consistent with ongoing positive selection (in the form of viral pathogens) over long expanses of evolutionary time. The TRIM5 loci of old world primates also display a high degree of within-species variation (polymorphism), a fact that can be exploited to study the biological manifestations of restriction in vivo. We have identified substitutions in the SIV capsid protein corresponding with emergence of SIVsm (as pathogenic SIVmac) after unintentional cross-species transmission into rhesus macaque colonies in the 1970s, and confirmed that these changes reflect adaptation to circumvent the species barrier presented by common alleles of rhesus macaque TRIM5. Additionally, retrospective analysis of macaque cohorts infected with SIV revealed a correlation between TRIM5 genotype and viremia, and in a subset of animals we tracked de novo emergence of TRIM5-resistant virus. Such results provide direct evidence that TRIM5 suppresses viral replication in vivo, thus exerting selective pressure during the earliest stages of cross-species transmission, and reveal that adaptation to overcome TRIM5 in the new host was a pre-requisite for emergence of pathogenic SIVmac in the 1970s. As a practical matter, the existence of functional polymorphism in TRIM5 has implications for both preclinical vaccine and pathogenesis studies conducted in nonhuman primate models of AIDS, and is a potential barrier to the development of a bonafide animal model of HIV infection.

SYMPOSIA

36

AIDS Vaccine 2011

Symposium 04: Integrated and Emerging Novel Issues in Clinical Trials S04.01
Passive Immunization to Prevent HIV Infection: Potential Applications and Challenges
B. Graham1
1

Invited Speakers

S04.02
Development of Ibalizumab and Improved Variants for Passive Immunization Against HIV
D. D. Ho1
1

Vaccine Research Center, NIH, NIAID, Bethesda, MD, USA

Aaron Diamond AIDS Research Center, New York, NY, USA

Diagnosis and treatment of HIV-infected persons is a highly effective method for preventing transmission to partners. However, thousands of new HIV infections occur each day at a higher rate than HIV-infected persons are entered into ARV treatment. Therefore, practical approaches to prevention are needed. Daily PrEP in uninfected persons at high risk can effectively prevent infection, but requires hundreds of persons to be compliant with daily treatment to prevent a single transmission event. Therefore, passive or active immunization approaches that can achieve longer periods of protection would be useful in future prevention efforts. Protection afforded by licensed anti-viral vaccines is based primarily on the induction of neutralizing antibody. Induction of antibodies with broadly neutralizing activity against commonly transmitted strains of HIV has been difficult to achieve. Recently, new technological approaches have resulted in the identification of multiple gp160-specific monoclonal antibodies (mAbs) with diverse specificities and broad neutralizing capacity. VRC01 is an IgG1 mAb that targets the CD4 binding site and can neutralize 91% of Tier 2 isolates with an EC50 of 50 g/ml and 72% at 1 g/ml. It does not react with normal human tissue or antiphospholipid activity. There are no unusual structural features of VRC01 and the CDR3 loop is of average length (13aa), but the variable regions are highly somatically mutated (30%) from germline. Passive administration of VRC01 to macaques can prevent infection from a mucosal SHIV challenge.The presentation will review clinical trial options to: 1) establish proof-of-concept that antibody elicited by vaccine antigens with VRC01-like properties could prevent HIV infection, 2) define the pharmacokinetics and feasibility of using VRC01 as passive prophylaxis alone or combined with other modalities, and 3) determine whether VRC01 has antiviral properties that may be useful therapeutically.

There is no effective vaccine to protect against HIV infection today, and none will be available for the foreseeable future. The lack of an effective HIV vaccine is in part due to the structural properties of the viral envelope glycoprotein, gp120, which possesses highly variable amino-acid sequences along with extensive glycosylation that shield the virus from neutralizing antibodies. As an alternative strategy, our group is pursuing the use of antibodies by passive administration for the prevention of HIV infection. Specifically, we are utilizing a humanized monoclonal antibody, ibalizumab, directed to domain 2 of human CD4 that potently blocks HIV infection without causing any evidence of immune impairment. In the setting of HIV therapy, ibalizumab has already been shown to be effective against HIV by lowering viral load by one log in patients. It is also well tolerated, with no severe adverse events in about 250 infected patients, some for up to 3 years. With support from the Bill & Melinda Gates Foundation, we are now pursuing the proof-of-concept studies with ibalizumab in a phase 1 study in healthy volunteers. A phase 2a study will be conducted in 2012, and a phase 2b efficacy trial is anticipated to begin in 2013 in heterosexuals in South Africa and/or homosexual men in China. In parallel, we are re-engineering the antibody to come up with a second-generation candidate that has enhanced potency and breadth. Through the creation of fusion antibodies, we have in hand a number of constructs with 100% breadth against a panel of 118 strains of HIV in vitro. Importantly, one of these constructs has a potency that is ~225-fold more potent than the parental ibalizumab. We are also modifying the Fc and Fab regions of the antibody in order to improve its pharmacokinetic profile, largely by decreasing endosomal degradation and increasing recycling. Lastly, we are exploring the use of gene transfer methods that could produce the desired HIV-blocking antibody in muscle cells in vivo. Early results in rodents suggest such an approach is feasible.

AIDS Vaccine 2011

37

SYMPOSIA

Invited Speakers
S04.03

Symposium 04: Integrated and Emerging Novel Issues in Clinical Trials S04.04
Regulatory Considerations for the Approval of Novel HIV Vaccine Clinical Trial Designs in a Developing Country
G. Steel1
1 Management Sciences for Health (MSH), Beacon Bay, South Africa

Challenges in the Design of Future Combination Prevention Trials


M. Sobieszczyk1
1

Columbia University, New York, NY, USA

Recent developments in the field have placed systemic and topical antiretrovirals for prevention in the spotlight, highlighting the importance of incorporating combination approaches into future clinical trials. Significant progress has been achieved in the arena of drugs for prevention, both as PrEP and as treatment of HIV infected persons, with promising results from studies like CAPRISA 004, iPrEx and HPTN 052. Relevance of ARTs for prevention will only increase as more data emerge about applicability to diverse populations, and optimal timing and frequency of dosing around exposure. In this rapidly evolving landscape, a multimodality combination approach to biomedical prevention is thus an increasingly important area of HIV clinical research. This talk will aim to highlight some of the opportunities and challenges associated with designing future vaccine clinical trials as the field adapts to and incorporates advances in nonvaccine biomedical interventions.

Researchers have proposed various initiates that impact upon the regulatory review and approval of HIV vaccine clinical trials in a developing country. This presentation will explore adaptive clinical trials from a regulators perspective and the approach to consideration of standard of care for the comparator arm and/ or the background intervention. Issues discussed will include the role of the pre-clinical findings, DSMBs, phase of development, and ethical considerations.

SYMPOSIA

38

AIDS Vaccine 2011

Symposium 04: Integrated and Emerging Novel Issues in Clinical Trials S04.05
Challenges of Integrating Mucosal Immune Assessments Into HIV Vaccine Trials
A. Omu Anzala1
1 Kenya AIDS Vaccine Initiative / University of Nairobi, Nairobi, Kenya

Invited Speakers

Majority of individuals worldwide get infected with HIV through mucosal surfaces. Antibody inducing and cell-mediated vaccine candidates have recently shown either modest or no efficacy against HIV acquisition and no efficacy against HIV viral load set point. Although the immune mechanism of protection remains poorly understood, new HIV vaccine candidates are now being designed at inducing protective mucosal immunity. Little has been done in the past to review mucosal sampling and develop appropriate standardized assays to assess vaccine-induced immune response. My research unit, Kenya AIDS Vaccine Initiative (KAVI), in collaboration with the International AIDS Vaccine Initiative (IAVI) and the University of Toronto and the University of Manitoba have jointly put together three protocols to study mucosal sampling techniques, determine volunteer acceptance of these sampling techniques, and develop and validate a number of mucosal assays that can be used in multiple sites to assess vaccine induced immunities. My presentation will cover our experiences in mucosal sampling, volunteer acceptance and assay development. I will also discuss the process KAVI is undertaking to establish itself as a leader in mucosal immunology.

AIDS Vaccine 2011

39

SYMPOSIA

Invited Speakers
S05.01
Systems Biology of Dengue Infection
M. Kwissa1, H. I. Nakaya1, B. Pulendran1
2

Symposium 05: Systems Biology S05.02


Whole Blood Transcriptional Monitoring of Acute HIV-1 Infection Reveals Differential Signatures of Host Immune Activation
J.A. Skinner1, N. Baldwin1, M. Sharma1, B. Lemoine1, D. Blankenship3, H. Qin3, Z. Xu1, E.B. DeVol3, V. Pascual1, A. Mejias4, O. Ramilo4, R. Pankla5, G. Lertmemongkolchai6, M. Berry7, A. OGarra7, K. Palucka1, R. Thiebaut8, G. Chene8, M. Cohen9, K. Soderberg10, T.N. Denny10, A. McMichael11, B.F. Haynes10, Y. Levy2, J. Banchereau1, D. Chaussabel1, NIAID Center for HIV/AIDS Vaccine Immunology1
1 Baylor Institute for Immunology Research, Dallas, TX, USA; 2Henri Mondor Hospital-ANRS, Creteil, France; 3Baylor Institute for Health Care Research and Improvement, Dallas, TX, USA; 4Nationwide Childrens Hospital Center for Vaccines and Immunity, Columbus, OH, USA; 5Department of Medical Technology, Naresuan University, Phitsanulok, Thailand; 6Department of Clinical Immunology, Khon Kaen University, Khon Kaen, Thailand; 7MRC National Institute for Medical Research, London, United Kingdom (Great Britain); 8INSERM U897, Bordeaux, France; 9University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; 10Duke University School of Medicine, Durham, NC, USA; 11Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom (Great Britain)

Emory Vaccine Center, Emory University, Atlanta, GA, USA

Applying systems biology tools to study immunological mechanisms of viral infection can greatly impact development of successful vaccines or drug treatments. We have undertaken a systems biological approach to assess the immunological and molecular mechanisms behind dengue infection in patients with different clinical outcomes. Dengue viruses (DENV) are emerging, mosquito-borne flaviviruses and the causative agents of dengue fever (DF) and dengue hemorrhagic fever (DHF). Millions of people are infected by DENV each year with several hundred thousand, especially in children, progressing to DHF, which is a life-threatening disease. Understanding early events occurring after infection is crucial for accurate diagnosis and determining appropriate treatment. Verifying early mechanisms of DENV invasion and interactions with host immune response will further allow development of a vaccine and antiviral drugs. We used an integrated approach involving, multi parametric analysis of innate and adaptive immune cells, serum cytokines and chemokines and gene expression profiles in a cohort of DENV infected patients in Bangkok, Thailand. Interestingly, we observed an expansion of blood innate cells including neutrophils, inflammatory dendritic cells and monocytes and activated NK cells in DF and DHF patients. Our blood transcriptome analysis identified several genes that correlated with clinical and immunological data, revealing pathways potentially relevant for the innate immunity against dengue infection. I will discuss a comprehensive data set on the human immune response to dengue infection with emphasis on early events governing subsequent adaptive immunity and clinical outcome.

SYMPOSIA

Genome-wide transcriptional studies in non-human primates have suggested that the quality and magnitude of early immune responses to SIV infection are associated with downstream disease progression. The aim of this study is to identify, monitor, and interpret the whole blood transcriptional signature of acute HIV-1 infection (AHI). A total of 56 AHI patients and matching healthy controls from both Africa and the United States were distributed into training, test, and validation datasets. Whole blood from these subjects was obtained for transcriptional profiling using microarrays. Both gene and modular transcriptional framework analyses were utilized to interpret the signature of AHI and for comparisons with other disease signatures including: influenza, tuberculosis, sepsis, and chronic HIV following interruption of anti-retroviral therapy (ART). Additional samples were collected at 1, 2, 4, 12, and 24 weeks post-enrollment for training and test set AHI subjects, to establish the longevity of the signature in the presence or absence therapy. We identified a robust AHI signature with increased activity in: interferon, cell cycle, cytotoxic, plasma cell, and B-cell modules amongst others. This activity was unique when compared to signatures obtained from patients with other infections. Only interferon and cell cycle activity was observed in both AHI subjects and chronically infected patients following interruption of therapy. Notably, 20% of AHI patients were observed to express a quiescent signature that clustered independent of the highly active signature described above. We found that these patients had significantly lower viral set-points (p=0.01) when compared to patients with the active signature. Finally, longitudinal analysis demonstrated that the active AHI signature can persist independent of viral load in ART treated patients and up to 6 months in the absence of therapy.Transcriptional monitoring of early HIV infection offers both quantitative and qualitative assessments patient responses that may be predictive of long-term disease progression.

40

AIDS Vaccine 2011

Symposium 05: Systems Biology S05. 03


Functional Genomic Analysis of the Human T Cell Response to Chronic Viral Infection
W. Haining1
1

Invited Speakers
S05.04
Correlates of Immune Protection: A System Biology Analysis of Vaccine and Natural Mediated Immune Protection
R. Sekaly1, F. Lefebvre1, A. Filali1, P. Wilkinson1, M. Caskey2, S. Schlessinger2, R. Steinman2, M. Cameron1
1

Dana-Farber Cancer Institute, Boston, MA, USA

We have used integrative genomic approaches to identify genes and pathways that impair T cell function in chronic viral infection in humans. Traditionally, immunologists have used flow cytometry and functional assays to define combinations of markers that are representative of specific lineages and functional states within the immune system. However genome-scale technologies now provide the ability to define with increasing resolution complex sets of markers expression signatures that represent discrete biological properties of cell populations. Because transcriptional signatures can serve as surrogates of a phenotype, function or cell state, they can integrate phenotypic information between experiments, cell-types and species. We have analyzed geneexpression profiles from HIV-specific CD8 T cells in patients with HIV and integrated these with transcriptional profiles from mouse models of chronic viral infection as well as in vitro experiments. We have used these orthogonal datasets to identify novel regulators of T cell function that would not have been apparent through the analysis of any single dataset in isolation. Integration of expression signatures across experimental conditions together with functional analysis of their component genes can therefore provide new opportunities to dissect the regulation of the adaptive immune response to chronic viral infection in humans.

Vaccine & Gene Therapy Institute of Florida, Port St Lucie, FL, USA; 2Rockefeller University, New York, NY, USA
The development of vaccines to HIV and other chronic viral infections has been considerably slowed down by the lack of licensed adjuvants capable of inducing a strong innate immune response that triggers the development of humoral and cellular immunity. The lack of correlates of immune protection has also been a major impediment to the development of a safe and efficacious vaccine. Approaches that can monitor the integrated immune response as compared to the evaluation of its individual components are essential to reach these two objectives namely the identification of adjuvants and correlates of immune mediated protection; we have used systems biology to monitor the innate immune response induced by an adjuvant known to induce a strong adaptive immune response in rodents and non-human primates; we show that this innate immune response has several features in common with that induced by a complex virus known to be a highly efficient vaccine, i.e. the Yellow Fever vaccine. The Interferon pathway clearly stood out as the major pathway induced by this adjuvant; other pathways of the innate immune response were also induced by both Poly ICLC and YF17D. Interestingly interferons have also been shown to be very much involved in the progression of several chronic viral infections including HIV and SIV; we will show that the Interferon signatures that are associated to protection are different than those induced during a disseminated viral infection. System biology and the meta-analysis of public databases is essential to the development of novel safe and efficacious adjuvants and to the identification of predictors of efficacious innate immune responses. This work was funded in part by grants to RPS from the Bill & Melinda Gates Foundation and from the NIH and to RMS from the NIH.

AIDS Vaccine 2011

41

SYMPOSIA

Invited Speakers
S05.05

Symposium 05: Systems Biology

Systems Vaccinology: Using the Tools of Systems Biology to Enable Rational Vaccine Design
A. Aderem1
1

Seattle Biomedical Research Institute, Seattle, WA, USA

Current approaches to making vaccines are similar to those used by Jenner more than 200 years ago in which a related, weakened or dead form of the infectious agent or a component of the pathogen is used to elicit immune memory. Some vaccines have been very successful and have resulted in the virtual eradication of scourges such as smallpox, diptheria, polio, and measles. Other vaccines have failed, most notably those against HIV, tuberculosis, and malaria. The current approach is essentially trial and error and has not been accompanied by a fundamental understanding of precisely how protection arises. This lack of understanding has hampered progress in the redesign of unsuccessful vaccines. Data will be presented that support the argument that Systems Biology offers a new approach to address the complexity of the immune system and will provide the tools for rational vaccine design. Systems approaches will also speed up vaccine trials, streamline and improve the manufacture of vaccines, and aid in field-testing.

SYMPOSIA

42

AIDS Vaccine 2011

Symposium 06: New Concepts in Protection Against HIV Acquisitions S06.01


Harnessing Antibody-Mucus Interactions to Prevent HIV Transmission
T. Hope1, S. Shukair1, S. Allen1, G. Cianci1
1

Invited Speakers

S06.02
a47/CD4 T Cells are Key Targets in Mucosal Transmission of HIV
J. Arthos1
1 National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA

Northwestern University, Chicago, IL, USA

Male-to-female transmission of human immunodeficiency virus type-1 (HIV-1) accounts for a majority of adult infections worldwide. During male-to-female transmission the virus must traverse a protective barrier of cervical and cervical-vaginal mucus before reaching protective mucosal epithelial barriers. Using the direct observation of viral particles and nanobeads within cervical mucus (CM) and cervical/vaginal mucus (CVM) we are able to directly quantify and analyze particle transport. The pH of every sample is recorded. Virions lacking envelope and nanobeads act as internal controls that accounts for any intraand inter-donor heterogeneity within the mucus. Individual virion diffusion was analyzed using custom computer-based particle trafficking software. We then compared the movement of HIV and controls in the presence or absence of different types of HIV binding and control antibodies. We find that the binding of antibodies to HIV causes a decrease in the transport of HIV within mucus. The change in transport is dependent on antibody binding as demonstrated by antibody binding specificity. Further, decreased transport was pH dependent and the pH dependence correlated with the pH sensitivity of a particular antibody in a virus capture assay. We have now identified multiple neutralizing and non-neutralizing anti-envelope antibodies that can decrease the mobility of the wild-type virus relative to the controls in CM and CVM. The presence of seminal plasma does not inhibit this mechanism of antibody action. Therefore, the presence of HIV antibodies within the female genital tract can sequester virus within mucus, potentially inhibiting an eventual interaction with a target cell that could mediate transmission. These results reveal a novel mechanism for anti-envelope antibodies to inhibit HIV transmission in women and have potentially important implications for vaccine development. They describe a mechanism whereby, broadly binding, but not broadly neutralizing antibodies to inhibit HIV sexual transmission.

Mucosal transmission of HIV is inefficient. The virus must breach physical barriers before it infects mucosal CD4 T cells. Low-level viral replication occurs initially in mucosal CD4 T cells, but within days high-level replication occurs in Peyers patches, mesenteric lymph nodes and the gut lamina propria. The viral quasispecies in a donor contracts through a genetic bottleneck in the recipient, such that infection is typically established by a single founder virus. Founder viruses in subtypes A and C mucosal transmission tend to encode gp120s with reduced numbers of N-linked glycosylation sites at specific positions throughout the V1- V4 domains, relative to typical chronically replicating isolates. The transmission advantage gained by the absence of these N-linked glycosylation sites is unknown.Using primary a47/CD4 T cells and a flow-cytometry based steady-state binding assay we have determined that the removal of transmission-associated N-linked glycosylation sites from chronic subtype A and C envelopes results in large increases in the specific reactivity of gp120 for integrin-a47. These results suggest that the genetic bottleneck reported at the time of transmission may frequently involve a requirement for the productive infection of a47/CD4 T cells. Understanding the structural features of the HIV envelope that promote reactivity with integrin a47 receptor may help define early events in HIV transmission.

AIDS Vaccine 2011

43

SYMPOSIA

Invited Speakers
S06.03

Symposium 06: New Concepts in Protection Against HIV Acquisitions S06.04


Defining the Spectrum of Vaccine Induced Humoral Responses
G. Tomaras1
1

Critical Role of Retinoic Acid and TLR Signals in Gut-Associated Dendritic Cell Differentiation
J. Mora1, E. J.Villablanca1, S. Wang1
1 Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA

Duke University, Durham, NC, USA

The intestinal mucosa confers protection against the myriad of potentially pathogenic microorganisms to which it is constantly exposed. Lymphocytes need the integrin a47 and the chemokine receptor CCR9 to efficiently home to the intestinal mucosa in order to confer immune protection in this compartment. We and others have shown that gut-associated dendritic cells (DC) from mesenteric lymph nodes, Peyers patches, and small intestine lamina propria, metabolize vitamin A into all-trans retinoic acid (RA), which is required for inducing a47CCR9 gut-tropic lymphocytes and for promoting the differentiation of gut-homing intestinal IgA antibody-secreting cells (IgAASC). Thus, gut-associated DC and RA shape gut mucosal immunity by modulating T and B cell migration and IgA-ASC differentiation.We recently showed that RA induces a positive feedback loop by stimulating its own synthesis in DC. RA was physiologically necessary and sufficient in vivo to confer gutassociated DC with the capacity to synthesize RA and to induce gut-tropic T cells and IgA-ASC. RA-mediated DC education needed ERK/MAPK signaling and, unexpectedly, required the intracellular adaptor MyD88, which is conventionally associated with TLR and IL-1/IL-18 signaling. Thus, our data indicate that RA and MyD88 are key mediators of DC education in the gut mucosa, uncovering a novel crosstalk between RA and MyD88dependent pathways (Villablanca et al., Gastroenterology, 141: 176, 2011; Wang et al., J. Immunol., 187: 141, 2011).In summary, our data indicate that RA and TLR signals intersect to educate DC to synthesize RA and to induce gut-tropic T and B cells and IgA-ASC. Our findings could offer a straightforward alternative to boost intestinal immune responses for vaccination purposes, such as during HIV infection, where the small intestine has been identified as a major reservoir for the virus.

The results of the heterologous prime-boost RV144 efficacy trial raised the hypothesis that a protective B cell response played an important role in reduced rates of virus acquisition. Moreover, specific antibody responses have been identified as the immune correlates of protection in several non-human primate studies. Analyses of HIV-1 specific IgA and nonneutralizing IgG antibodies will be discussed in the context of which antibody types and specificities are likely central to a protective immune response.

SYMPOSIA

44

AIDS Vaccine 2011

Symposium 06: New Concepts in Protection Against HIV Acquisitions S06.05


Correlates of Protection Against SIVmac251 in Vaccinated Rhesus Monkeys
D. Barouch1
1

Invited Speakers

Beth Israel Deaconess Medical Center, Boston, MA, USA

Defining the immunologic correlates of protection against HIV1 represents a major challenge for HIV-1 vaccine development. Given the inability of HIV-1-infected individuals to clear virus, immunologic correlates need to be elucidated in the context of vaccine efficacy trials. In two preclinical vaccine studies, we show that there are distinct correlates of protection against acquisition of infection and virologic control following heterologous challenge of vaccinated rhesus monkeys with the neutralizationresistant virus SIVmac251.

AIDS Vaccine 2011

45

SYMPOSIA

Invited Speakers
S07.01

Symposium 07: Characterization of Breakthrough Viruses S07.02


Sequence Analysis of HIV-1 Breakthrough Infections in the RV144 Trial
M. Rolland1, B. Larsen2, P. Edlefsen3
1 U.S. Military HIV Research Program, Henry Jackson Foundation, Rockville, MD, USA; 2University of Washington, Seattle, WA, USA,3SCHARP/VIDI, Seattle, WA, USA

A Deeper Look at The Virus Populations Infecting Step Trial Volunteers


S. Iyer1, H. Bouzek1, M. Kim1, B. Larsen1, E. Casey1, M. Rolland2, P. Edlefsen3, T. Hertz3, C. Magaret3, N. Frahm3, D. Westfall1, K. Wong1, H. Zhao1, E. Sanders-Buell2, S. Tovanabutra2, J. N. Stoddard1, S. Nariya1, P. Konopa1, B.S. Maust1, W. Deng1, A. Bates2, A. Bradfield2, M. Bose2, A. OSullivan2, J. Hural3, D.R. Casimiro4, M.N. Robertson4, N. Michael2, M.J. McElrath3, L. Corey3, S. Buchbinder5, P. B. Gilbert3, J. Kim2, J.I. Mullins1 University of Washington, Seattle, WA, USA; 2U.S. Military HIV Research Program, Rockville, MD, USA; 3Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 4Merck Research Laboratories, West Point, PA, USA; 5San Francisco Department of Health, San Francisco, CA, USA
1

In the first analysis of virus breakthrough sequences from the STEP trial, we found evidence that the viral populations infecting vaccinees were depleted in epitopes present in the vaccine (Rolland et al, Nat. Med. 17:366 (11)). This result implies that vaccine-induced immunity within the host could block the outgrowth of strains with epitopes homologous to the vaccine, or, promote emergence of escape variants that evolved quickly within the newly infected host. To distinguish between the two above hypotheses we are deeply sequencing (at least 1,000-5,000 viral templates using pyrosequencing technology) virus populations from the earliest time points available, to search for evidence of pre-escape mutants. Using the same techniques, we are following early evolutionary events in these viral populations over the first year of infection to test the hypothesis that immunological escape, evidenced by viral population changes within or near predicted and defined epitopes, will occur earlier or be more robust in vaccinees than placebo recipients. This would be interpreted as reflecting an anamnestic response resulting from prior exposure to the vaccine. Longitudinal sequence data will also facilitate a more precise and statistically powerful analysis of potential sieve effects on the pre-seroconversion virus.

The positive results of the RV144 trial, which showed modest and transient protection from HIV-1 infection in Thai volunteers, prompted us to test for a sieve effect on breakthrough sequences. We characterized 1,029 near-full-length HIV-1 genome sequences obtained at HIV-1 diagnosis from 50 vaccine and 71 placebo recipients. All but ten volunteers were infected with CRF01-AE viruses.We found heterogeneous founder viral populations in 33% of vaccinees and 38% of placebo recipients (p = 0.56). The mean pairwise intra-host sequence diversity was on average 0.50% in env, a high number reflecting the fact that most individuals were diagnosed in Fiebig stage 5-6. There was a weak relationship between the env diversity and the time since the last HIV-1 negative visit: Spearman Rhos coefficient ? = 0.26 (p = 0.005).To test for a sieve effect on breakthrough sequences, we performed a series of local, global and Bayesian pre-specified tests. Among these, we compared how the founder sequences from individuals (infected with CRF01-AE) diverged from the vaccine strains. We found that Env sequences from vaccinees (0.207) tended to be more divergent from the CRF01AE vaccine sequence than those from placebo recipients (0.189), yet this difference was not significant (p = 0.16). We identified a set of signature sites associated with vaccination in the V2 loop, a finding bolstered by the focus of RV144 vaccine-induced immune responses on V2, a region of Env not typically targeted by immune responses in natural infection.Our results showed that sampling at 6-month intervals failed to identify most infections at the acute stage, and illustrated how intra-host evolution can confound analyses of founder viral populations, emphasizing the importance of frequent sampling in future vaccine trials. Together with immunological data pointing to V2, our sequence analyses provide a possible mechanistic factor behind the protection conferred by the RV144 vaccine combination.

SYMPOSIA

46

AIDS Vaccine 2011

Symposium 07: Characterization of Breakthrough Viruses S07.03


Increased Multiple Variant Transmission in VAX003 Injection Drug Users
K. Bar , S. Sterrett , B. Hahn , G. Shaw
1 1 1 1 1

Invited Speakers

S07.04
Sieve Analysis of RV144
P. Edlefsen1, T. Hertz2, C. Magaret2, A. DeCamp2, P. Gilbert2
1 Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 2SCHARP at the Fred Hutchinson Cancer Research Center in Seattle, WA, USA

University of Alabama at Birmingham, Birmingham, AL, USA

Background: VAX003 was a HIV-1 vaccine efficacy trial of four doses of a recombinant gp120 (AIDSVAX) subtype B/E conducted in a cohort of injection drug users (IDUs) in Bangkok, Thailand that demonstrated no efficacy. To explore HIV transmission in IDUs and the role of HIV acquisition risk factor in vaccine studies, we are performing transmitted/founder virus analyses on the HIV-infected IDUs participants of VAX003. Methods: Plasma from all HIV-infected VAX003 subjects with samples obtained within 100 days of their estimated date of infection (n=18 in placebo arm, n=32 in vaccine arm) was obtained from Global Solutions for Infectious Diseases. Western blots for clinical staging, plasma RNA extraction/cDNA synthesis and single genome amplification of full env genes were performed. Sequences were manually aligned, displayed in phylogenetic trees and Highlighter plots, and analyzed within a model of neutral viral evolution. Results and Conclusions: In the placebo arm, a total of 704 env sequences were generated (19-36 per subject time point, median 30). The frequency of multiple variant transmission (MVT) in the placebo recipients was 44% (8/18), which is significantly higher (p=0.028, Fishers exact test) than the 19% MVT rate in sexual transmission cohorts (Keele, et al 2008, Abrahams et al, 2009, Haaland et al, 2009). Sequencing of the vaccine arm is ongoing, but preliminary analyses indicate 21% MVT (3/14), with an additional 18 subjects yet to process. Work is ongoing to complete the sequence analysis, relate any differences in transmission to neutralizing antibody responses, and determine if the study vaccine candidate may have produced a subtle vaccine response that affected the rate of MVT, but did not rise the level of protection from HIV acquisition.

A presentation of initial results from SCHARPs pre-specified analyses of the HIV genome sequences obtained from infected participants of the RV144 trial for the identification of sieve effects and of post-infection CTL escape.

AIDS Vaccine 2011

47

SYMPOSIA

Invited Speakers
S07.05

Symposium 07: Characterization of Breakthrough Viruses

Understanding HIV Transmission Utilizing Transmitted/Founder Viruses


B. Keele1
1

SAIC-Frederick, National Cancer Institute, Frederick, MD, USA

Genetic diversity is significantly reduced from donor to recipient during HIV-1 transmission. For the vast majority of mucosal transmissions this genetic bottleneck is so severe that only a single individual transmitted lineage can be identified in blood plasma during acute infection. Generating infectious molecular clones from various transmitted viruses has confirmed CCR5 utilization with a general lack of macrophage tropism. Additionally, nonhuman primate models have been developed to more precisely mimic the essential features of HIV-1 transmission. Utilizing both SIVmac251 and SIVsmE660 isolates, protocols and inoculum concentrations have been developed for rectal, vaginal and penile infections where most animals were infected with one or few viral variants. Again, infectious molecular clones were generated from these transmitted SIV variants. As with HIV, each clone was infectious and replication competent. Reintroduction of each clone into naive rhesus macaques revealed similar replication profiles and pathology seen in the initial animals. Future use of molecularly defined, pooled clonal viruses could be useful in elucidating the earliest events in viral transmission and could improve consistency and reproducibility of vaccine efficacy trials in NHPs.

SYMPOSIA

48

AIDS Vaccine 2011

Oral Abstract Session 01: Vaccines and Other Prevention Strategies OA01.01
DNA/NYVAC Vaccine Regimen Induces HIVSpecific CD4 and CD8 T-Cell Responses in Intestinal Mucosa
M. Perreau1, H.C. Welles1, A. Harari1, O. Hall1, R. Martin1, M. Maillard2, G. Dorta2, P. Bart1, E.J. Kremer3, J. Tartaglia4, R. Wagner5, M. Esteban6, Y. Levy7, G. Pantaleo1 Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland; 2Centre Hospitalier Universitaire Vaudois (CHUV)/Gastroenterology, Lausanne, Switzerland; 3Institut de Gntique Molculaire de Montpellier (IGMM), Montpellier, France; 4sanofi pasteur, Swiftwater, PA, USA; 5Institute of Medical Microbiology, University of Regensburg, Regensburg, Germany; 6Department of Molecular and Cellular Biology (CNB-CSIC), Madrid, Spain; 7Institut National de la Sant et de la Recherche Mdicale, Unit U955, Crteil, France
1

Oral Abstract Sessions

OA01.02
Cell-Mediated Immune Responses After DNA Delivered by Either Biojector or Electroporation and Boosted with a Heterologous Insert Recombinant Poxvirus
J.R. Currier1, S. Ratto-Kim1, V. Ngauy2, J. Ake1, C. Lau1, T. Doris1, E. Herrera1, W.B. David3, N.Y. Sardesai4, J. Boyer3, A.S. Khan4, J. Lee4, M. Bagarazzi4, J. Yan4, E. Adams5, P. Earl6, B. Moss6, J. Kim1, N. Michael1, M. Robb1, M.A. Marovich1
1

Military HIV Research Program, Rockville, MD, USA; 2Armed Forces Research Institute for Medical Sciences, Bangkok, Thailand; 3University of Pennsylvania School of Medicine, Philadelphia, PA, USA; 4Inovio Pharmaceuticals, Blue Bell, PA, USA; 5Divisions of AIDS, NIAID, Bethesda, MD, USA; 6 Laboratory of Viral Diseases, NIAID, Bethesda, MD, USA
Background: We conducted a phase I, randomized, open label trial of PENNVAXTM-G DNA and MVA-CMDR to evaluate safety and immunogenicity of a heterologous DNA prime/recombinant poxvirus boost in human volunteers in the US prior to expansion of the trial into east Africa.

Background: HIV vaccine-candidates based on rare adenovirus serotypes such as Ad26 and Ad35 vectors, and poxvirus vectors are important components of future promising vaccine regimens that in the near future hopefully will move into a number of efficacy clinical trials in combination with protein vaccines. For these reasons, it is important to comprehensively characterize the vaccine-induced immune responses in different anatomical compartments and particularly at mucosal sites which represent the primary port of entry for HIV. Methods: In the present study, we have investigated the anatomic distribution in blood and gut mucosal tissues (rectum and ileum) of memory poxvirus-specific CD4 and CD8 T cells in subjects vaccinated with smallpox and compared with vector (NYVAC)specific and HIV insert-specific T-cell responses induced by an experimental DNA-C/NYVAC-C vaccine regimen. Results: Smallpox-specific CD4 T-cell responses were present in the blood of 52% of subject studied, while Smallpox-specific CD8 T cells were rarely detected (12%). With one exception, Smallpox-specific T cells were not measurable in gut tissues. Interestingly, NYVAC vector-specific and HIV-specific CD4 and CD8 T-cell responses were detected in almost 100% of the subjects immunized with DNA-C/NYVAC-C in blood and gut tissues. The large majority (83%) of NYVAC-specific CD4 T cells expressed a4b7 integrins and the HIV co-receptor CCR5. Conclusion: These results demonstrate that the experimental DNA-C/NYVAC-C HIV vaccine regimen induces the homing of potentially protective HIV-specific CD4 and CD8 T cells in the gut, the port of entry of HIV and one of the major sites for HIV spreading and depletion of CD4 T cells.

Results: Early analysis of the ICS assay data has revealed that a strong cell-mediated immune response was generated in subjects receiving either the BioJector or Electroporation delivered DNA and a rMVA-CMDR boost. IFN producing CD4+ and CD8+ T cells specific for both Gag and Env peptide sets matching the rMVACMDR inserts were detected at frequencies as high as 0.57% (Envspecific CD8+ T cells) and 0.37% (Env-specific CD4+ T cells). The phenotype of the responding cells has been identified as effectormemory cells (CD45RO+/CCR7-/CD28-) for both the CD4+ and CD8+ T cell populations. The highest magnitude responses were seen after the MVA-CMDR boost. Conclusion: PENNVAXTM-G DNA prime/MVA-CMDR boost generates a balanced CD4+/CD8+ HIV-specific T cell-mediated immune response. Elispot and further ICS assays using DNA insert matched peptide sets are planned to determine the frequency of antigen-specific T cells after the DNA prime.

AIDS Vaccine 2011

49

ORAL ABSTRACT SESSIONS

Methods: Volunteers were given a priming vaccination with PENNVAXTM-G DNA at months 0 and 1, followed by boosting with MVA-CMDR at months 3 and 6. Immunomonitoring was performed at pre-immunization, and at 2 weeks post-DNA and MVA vaccinations respectively. The phenotype and function of HIV-specific T cells was examined by flow cytometry measuring IFN, IL-2, TNF, MIP-1 and CD107a. Simultaneous phenotyping for effector cell, effector-memory cell, and central memory cell populations was performed using the surface markers CD45RA, CD45RO, CCR7 and CD28.

Oral Abstract Sessions


OA01.03
Titered Mucosal Challenge of Rhesus Macaques with SIVmac251 Recapitulates HIV Vaccine Efficacy in Humans
G. Franchini1, P. Pegu1, M. Vaccari1, S. Gordon1, B. Keele2, M. Doster1, Y. Guan3, G. Ferrari4, D. Montefiori4, D. Venzon5, C. Fenizia1, J. Lifson6, N. Michael7, J. Kim7, J. Tartaglia8

Oral Abstract Session 01: Vaccines and Other Prevention Strategies OA01.04
Preservation of HIV-1-Specific CD4+ T-IFN-Cell Responses in Intercurrent Infections Following Exposure to Tenofovir Gel
M. Mureithi 3, D. Poole3, V. Naranbhai1, S. Reddy2, N.P. Mkhwanazi2, S. Sibeko1, Q. Abdool Karim1, S. abdool Karim1, T. Ndungu2, M. Altfeld3, CAPRISA 004 Trial Group1 Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa; 2HIV Pathogenesis Programme, Doris Duke Medical Research Institute, South Africa; 3University of Kwazulu-Natal, Durban, South Africa
1

NCI/NIH, Bethesda, MD, USA; 2AIDS and Cancer Virus Program NCI-Frederick, MD, USA; 3Human Monoclonal Antibody Core Laboratory, University of Maryland, MD,USA; 4Duke University Medical Center, Durham, NC, USA; 5Biostatistics and Data Management Section, NCI/NIH, MD, USA; 6Human Retrovirus Section, NCI/NIH, MD, USA; 7Walter Reed Army Institute of Research, MD, USA; 8sanofi pasteur, Inc, PA, USA
1

Background: A combination vaccine based on the recombinant canarypox vector ALVAC-HIV and HIV gp120 envelope glycoprotein protected nearly one third of vaccinees from HIV acquisition but afforded no protection from CD4+T-cell loss or high virus levels in vaccines that became infected in the RV144 trial in Thailand. This result was surprising, given the limited ability of either vaccine component to induce CD8+T-cell responses or elicit broadly neutralizing antibodies. An animal model able to predict HIV vaccine efficacy for humans could hasten progress in our understanding of the immune responses that contribute to protection. Methods: We vaccinated Indian rhesus macaques with an ALVACSIV and SIVgp120 immunization regimen that mimics the RV144 trial. At the end of the immunization regimen, the vaccinated animals received a titered mucosal challenge with a dose of SIVmac251 shown to transmit a limited number of variants to nave controls, recapitulating human mucosal HIV transmission. Results: Vaccine immunogenicity and efficacy in macaques were similar to observations in humans; as in RV144, one third of vaccinated macaques were protected from acquisition SIVmac251. Viral load and CD4+ T cell numbers in animals that did become infected were not different from controls. Animals protected from infection had equivalent T-cell responses but higher avidity binding antibodies to gp120 compared to unprotected animals. Conclusion: Titered intrarectal challenge of macaques with SIVmac251 shows potential to accurately model clinically observed vaccine efficacy, suggesting a role for this model in understanding protective responses elicited by HIV preventive vaccines. Consistent with the results from RV144, our findings suggest non-neutralizing antibody responses may play a role in protection from SIV/HIV acquisition.

Background: The CAPRISA004 microbicide trial demonstrated reduction of sexual HIV-1 acquisition in women using a vaginal microbicide gel containing tenofovir. Future vaccine development efforts need to factor in the consequences of prophylactic use of anti-retrovirals in the evolution of immune responses during intercurrent HIV-1 infection. From the microbicide trial, we compared innate and adaptive immune responses in women with primary/intercurrent HIV-1 infection following exposure to 1% tenofovir gel or placebo gel. Methods: Natural killer (NK) cells and myeloid dendritic cells (mDCs) frequencies and activation status, and HIV-1-specific T cell responses were cross-sectionally assessed using multi-parametric flow cytometry in 36 randomly selected HIV-1 clade C infected female adults exposed either to tenofovir microbicide gel (n=17) or placebo (n=19) at an estimated 3 months post HIV acquisition. Both the 17 tenofovir-arm and 19 placebo-arm women had similar CD4+ T cell counts and HIV-1 viral loads at time of change in serostatus as well as at the time of this study. Results: Overall, NK cell and mDCs frequencies and activation, and HIV-1-specific CD8+ T cell responses did not differ significantly between the two groups. In contrast, HIV-1-specific Gag IFN CD4+ T-cell responses were significantly higher (p = 0.01) in infected women randomized to the tenofovir arm. Conclusion: These data suggest that the use of tenofovir gel by women around the time of breakthrough HIV infection may lead to preservation of HIV-1-specific IFN CD4+ T cells. Overall, the use of tenofovir containing gel around the time of infection can modulate HIV-1 immunity, and these immunological changes need to be considered in future trials combining HIV-1 vaccines and microbicides.

ORAL ABSTRACT SESSIONS

50

AIDS Vaccine 2011

Oral Abstract Session 01: Vaccines and Other Prevention Strategies OA01.05
Homologous Priming and Boosting with Recombinant MVA, Prevention of Infection as Good as With the Heterologous Regimen of DNA Priming and MVA Boosting
H.L. Robinson , L. Lai , S. Kwa , P.A. Kozlowski , V. Hirsch , B.K. Felber5, G.N. Pavlakis5, P.L. Earl4, B. Moss4, R.R. Amara6
1 2 2 3 4 1

Oral Abstract Sessions

OA01.06 LB
Extended Evaluation of Volunteers Who Become HIV-1 Infected During Participation in a Phase III Vaccine Trial of ALVAC-HIV and AIDSVAX B/E
S. Rerks-Ngarm1, R.M. Paris2, S. Chunsutthiwat1, N. Premsri1, C. Namwat1, J. Bowornwatanuwong3, S. Li4, J. Kaewkungkal5, R. Trichavaroj6, M. de Souza7, D. Francis8, E. Adams9, S. Gurunathan10, J. Tartaglia10, R. OConnell2, C. Eamsila11, S. Nitayaphan11, V. Ngauy7, P. Thongcharoen12, P. Kunasol1, N. Michael13, M. Robb2, P. Gilbert4, J. Kim2 Prime-boost HIV Vaccine Phase III Trial, Nonthaburi, Thailand; 2US Military HIV Research Program, Rockville, MD, USA; 3Chonburi Regional Hospital, Chonburi, Thailand; 4 SCHARP, Seattle, WA, USA; 5Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; 6Department of Retrovirology, USAMC-AFRIMS, Bangkok, Thailand; 7MHRP/ AFRIMS, Bangkok, Thailand; 8GSID, San Francisco, CA, USA; 9 NIAID, NIH, Bethesda, MD, USA; 10sanofi pasteur, Swiftwater, PA, USA; 11Royal Thai Army, AFRIMS, Bangkok, Thailand; 12 Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; 13US Military HIV Research Program, Rockville, MD, USA
1

GeoVax Inc., Smyrna, GA, USA; 2Yerkes National Primate Research Center, Atlanta, GA,USA; 3 Louisiana State University Health Sciences Center, New Orleans, LA, USA; 4 National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA; 5National Cancer Institute at Frederick, Frederick, MD, USA; 6Vaccine Research Center, Emory University, Atlanta, GA, USA
Background: Heterologous priming and boosting has become popular for HIV/AIDS vaccine development. Here we report that a simpler homologous regimen of MVA priming and boosting elicits rates of prevention of rectal infection as good as those elicited by the more complicated heterologous regimen of DNA priming and MVA boosting. Methods: SIV239 vaccines produced non-infectious viruslike-particles (VLP) displaying trimeric membrane-bound Env. A DDMM regimen delivered DNA (3mg/i.m.) at 0 and 8 weeks and MVA (1x108 plaque forming units/i.m.) at 16 and 24 weeks. An MMM regimen delivered MVA at weeks 0, 8 and 24. Twelve weekly intrarectal challenges with 5000 50% tissue-culture infectious doses of SIVsmE660 were administered at 6 months following the final immunization. Results: The DDMM regimen elicited CD4+ T cells with higher breadth, magnitude, and degree of polyfunctionality than the MMM regimen. The MMM regimen elicited higher titer and higher avidity Env-specific IgG,higher ADCC activity, and higher Env-specific IgA responses in rectal secretions. Magnitudes, breadths and functionality of elicited CD8+ T cells and titers of neutralizing antibody were overall similar between groups. The best protective responses were in the MMM group. Following 6 challenges, 5 out of 8 MMM vaccinated animals (60%), 3 out of 8 DDMM vaccinated (40%) and 1 out of 23 unvaccinated controls (4%) remained uninfected. By 12 challenges, all unvaccinated animals were infected, whereas 2 out of 8 rhesus in both DDMM and MMM groups remained protected. The challenge infected 30% of the unvaccinated rhesus at each exposure, a much higher transmission rate than observed for typical human infections. Conclusion: Our results reveal the simpler MMM regimen eliciting more antibody and comparable, if not better, protection than the more complex DDMM regimen. Our results suggest that homologous priming and boosting with VLP expressing MVA immunogens may merit testing in Phase 2b efficacy trials.

Methods: Volunteers who became HIV infected in RV 144 were followed every 3 months with clinical and CD4 and HIV-1 viral RNA assessments. Both study volunteers and investigators remained blinded over the entire study period. Volunteers received HIV care and treatment according to the Thai Ministry of Public Health National Guidelines. Both modified Intent-to- Treat (mITT) and per-protocol (PP) analyses were performed using a time-toevent model based on a Cox proportional hazards. Secondary analyses of biomarker-based endpoints were assessed using marginal mean models fit by Generalized Estimating Equations and linear mixed models. Results: There were 48 PP and 61 mITT events among the 120 enrolled volunteers at the pre-specified, event-driven stopping point. Conclusion: Results of the study will be presented and discussed to include estimates of vaccine efficacy for disease progression as well as longitudinal comparison of biomarkers.

AIDS Vaccine 2011

51

ORAL ABSTRACT SESSIONS

Background: The Thai Phase III trial of ALVAC-HIV and AIDSVAX B/E was the first study to show modest efficacy for prevention of HIV infection in a community-based cohort. Of the 16,402 volunteers enrolled in the study, 132 became HIV-infected during the trial and 120 of these volunteers agreed to participate in RV152 to evaluate the effect of this prime-boost regimen on clinical disease progression as well as immunologic and virologic outcomes. The primary objective of the study was to evaluate differences among vaccine and placebo recipients in a composite endpoint comprised of clinical (AIDS-defining events, initiation of antiretroviral therapy) and laboratory (CD4-count <350 cells/ microliter) events.

Oral Abstract Sessions


OA02.01
Innate Immune Response Signatures to Ad5 (HVTN 071) and MVA (HVTN 205/908) Vectored Candidate HIV Vaccines Predict Induction of Adaptive Responses
E. Andersen-Nissen , D.E. Zak , A.C. Duerr , J.T. Chang , E.R. Peterson1, A.T. Krishnamurty1, T.R. Hensley1, D.J. Adams1, M.K. Hamilton2, J. Li2, A. Sato1, C.A. Morgan1, M.L. Elizaga1, S.C. DeRosa1, P.A. Goepfert3, H.L. Robinson4, A. Aderem2, M.J. McElrath1
1 2 1 1

Oral Abstract Session 02: Innate Immunity OA02.02


Frequent and Strong NK Responses to HIV-1 Env Peptides in Individuals with Acute and Chronic HIV-1 Infection
C.F. Thobakgale1, L. Fadda2, K. Lane2, F. Pereyra2, B.D. Walker2, T. Allen2, M. Altfeld2 University of KwaZulu-Natal, Durban, South Africa; 2Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA
1

Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Institute for Systems Biology, Seattle, WA, USA; 3University of Alabama at Birmingham, Birmingham, AL, USA; 4GeoVax Labs Inc., Smyrna, GA, USA
1 2

Background: NK cells play a critical role in the control of HIV-1 infection and NK cells that respond to HIV peptides have been recently described (Tiemessen et al, JI, 2009). However, the mechanisms by which NK cells recognize HIV-1 antigen are not understood. We investigated the frequency of NK responses to HIV peptide pools during acute and chronic HIV clade B infection. Methods: Individuals with acute HIV-1 infection (n=15), progressive chronic (n=10) and controlled chronic HIV-1 infection (n=10) were studied. Whole blood was stimulated with HIV peptide pools spanning the entire clade B consensus sequence and NK responses were quantified using multiparameter flow cytometry. Results: No NK cell responses to HIV-1 peptides were detected in HIV uninfected individuals. In contrast, 60% of chronically infected individuals and 58% of individuals with acute HIV1 infection had detectable IFN-+ NK cell responses to HIV-1 antigen (p=0.002 compared to negative individuals). IFN- and TNF- producing NK cell responses most frequently targeted Env (ranging from 2%-30% of all NK cells). Other HIV-1 proteins such as Gag, Pol and Nef were rarely targeted. Env-specific NK cell responses were fine-mapped to individual 18mer peptides and did not depend on HIV-1 specific T cell responses. In contrast, Env-specific T cell responses were most commonly absent in individuals with NK cell responses to Env. Conclusion: NK cells from HIV-1-infected individuals can respond frequently and strongly to HIV-1 peptides in vitro. The receptor/ligand interactions responsible for the recognition of HIV-1 require further investigation to rationally modulate NK cell responses to HIV-1.

Background: Identifying human innate immune signatures that predict adaptive immune responses may enable development of more efficacious HIV vaccines. Methods: We compared the innate immune responses after vaccination with either Ad5/HIV (MRKAd5 HIV-1 gag/pol/nef, n=35), or a DNA/HIV prime, MVA/HIV boost regimen (pGA2/JS7 DNA, MVA/HIV62 gag/pol/env, n=22) and identified signatures predictive of T-cell responses to HIV inserts. Results: No changes in serum cytokines, circulating leukocyte populations, or PBMC gene transcription were detected at 24 hours after DNA/HIV vaccination. In contrast, vaccination with Ad5/ HIV or MVA/HIV caused a decrease in circulating lymphocytes at 24 hours and an increase in CD14+CD16+ transitional monocytes at 72 hours post-vaccination. Profiling of 74 soluble factors in the serum revealed that Ad5/HIV induced changes in 21 while MVA/HIV altered only 8 (Hochberg p<0.05). Commonly induced factors included chemokines likely responsible for the alterations in circulating leukocytes such as IP-10 and MCP-1, as well as TNF-, IFN-, and TRAIL. Ad5/HIV showed higher magnitude induction of TRAIL, I-TAC, and MCP-2 than MVA/HIV and significantly induced the immunoregulatory cytokine IL-10. As seen for serum cytokines, PBMC transcriptional responses induced by MVA/HIV, including many interferon-inducible networks, were a subset of Ad5/HIV-induced responses. Ad5/ HIV vaccination uniquely up-regulated expression of 244 genes in PBMC, including C2, CD40, and IL-10. Linear discriminant analysis examining signatures common to the two vaccines revealed that serum changes in IP-10 and TRAIL at 24 hours post-vaccination predicted the induction 2-4 weeks later of Gagspecific CD8+ T-cells expressing IL-2 and/or IFN- with 75-90% accuracy. Analysis of transcriptional response signatures that predict T-cell and nAb responses is on-going. Conclusion: These data may help to explain differential induction and potency of adaptive immune responses by these two vaccine vectors, and such an approach can be applied to optimize future vaccine regimens.

ORAL ABSTRACT SESSIONS

52

AIDS Vaccine 2011

Oral Abstract Session 02: Innate Immunity OA02.03


NKp46 and NKG2D Receptors Are Implicated in the Priming and Suppression of HIV Replication by Natural Killer Cells Stimulated by MVA/HIV ANRS Vaccine
J. Cummings2, V. Arnold2, C. Didier2, A. Gilbert2, K. Yarbrough2, Y. Levy1, F. Barr-Sinoussi2, D. Scott-Algara2
1

Oral Abstract Sessions

OA02.04
HIV Infection Eliminates the Antibody-Dependent Cellular Cytotoxicity (ADCC) Functional Advantage of KIR3DL1-Expressing NK Cells from HLA-Bw4 Carriers
M.S. Parsons1, L. Wren2, G. Isitman 2, M. Navis2, I. Stratov2, N.F. Bernard1, S.J. Kent2
1

AP-HP, Groupe Henri-Mondor Albert-Chenevier, Immunologie Clinique, Crteil, France; 2Institut Pasteur, Paris, France

McGill University, Montreal, Canada; 2University of Melbourne, Melbourne, Australia

Background: Innate mechanisms are critical for the normal development of host immune responses to antigen. The interaction between Natural Killer (NK) and Dendritic cells (DC) is expected to greatly impact the establishment of both innate and adaptive immune responses to vaccine Methods: We describe in vitro responses of NK cells to recombinant Modified Vaccinia Virus Ankara expressing HIV-1 peptides (rMVA) compared to wild type vector (wtMVA) using an autologous in vitro co-culture system to analyze the NK response to DC exposed to the viral vector Results: Monocyte-Derived DC were efficiently infected with the rMVA virus and produced the encoded HIV-1 proteins. This being a cytopathic virus, we subsequently used a fluorescent dye system, to demonstrate that rMVA-infected DC were phagocytosed by uninfected DC after 48h in co-culture, allowing for cross-presentation in our in vitro system. MDDC infected with rMVA or the wtMVA control did not differ in their production of cytokines; and when these DC were cultured with autologous NK cells, they stimulated similar levels of NK proliferation. Both MVA strains were capable of inducing NK repertoire modifications, but comparative analysis indicated that NK stimulated with rMVA expressed higher levels of NKp30 and NKG2D. NK cells also showed increased degranulation when stimulated with rMVA-infected DC compared to wtMVA. This stimulation was contact-dependent and augmented when NKp46 was blocked during the NK:DCrMVA interaction. Functionally, rMVA-infected DC stimulated NK cells were better at subsequently suppressing HIV-1 in vitro as compared to wtMVA-stimulated cells. Blocking experiments showed NKG2D as major factor in this NK control of HIV-1 infection. NK cell stimulation by rMVA was specific since no differences were observed in CMV replication suppression between rMVA and wtMVA-stimulated NK cells Conclusion: These data demonstrate that recombinant MVA vaccine against HIV induced activated NK cells capable of specifically controlling HIV infection in vitro.

Background: Mechanisms for harvesting ADCC responses to develop better HIV vaccines are unclear. KIR3DL1 and HLA-Bw4 allelic combinations are associated with protection from HIV/ AIDS. The KIR3DL1/HLA-Bw4 combination facilitates licensing of NK cells, potentially enhancing killing of HIV-infected cells. Licensed KIR3DL1+ NK cells are, however, non-responsive to autologous HIV-infected cells, suggesting mechanisms other than direct killing are critical. We hypothesized that KIR3DL1+ NK cells from individuals co-carrying the HLA-Bw4 ligand overcome inhibitory signals and mediate enhanced anti-HIV ADCC. Methods: Blood from 21 healthy controls and 22 HIV-infected individuals was tested using an intracellular cytokine staining assay to detect NK cell activation in response to HIV-specific ADCC antibodies. Whole-blood was incubated for 5hr with HIV+ plasma and HIV envelope peptides and lymphocytes were stained to identify CD107a and IFN- expression from KIR3DL1+/- NK cells. Results: NK cells mediated robust ADCC responses. In HLA-Bw4+ individuals, KIR3DL1+ NK cells mediated anti-HIV ADCC, despite the inhibitory potential of the interaction between KIR3DL1 and HLABw4. NK cells from healthy control HLA-Bw4+ subjects exhibited higher ADCC bi-functionality in KIR3DL1+ than KIR3DL1- NK cells (16.5% vs. 11.6%; p<0.01). However, HIV-infected HLA-Bw4+ subjects demonstrated equal bi-functionity between KIR3DL1+ and KIR3DL1- NK cells (10.3% vs. 9.8%; p=0.7). Conclusion: KIR3DL1+ NK cells, from HLA-Bw4+ healthy controls exhibit robust anti-HIV ADCC, and represent a more functional subset than KIR3DL1- NK cells. This advantage is lost in HIVinfected individuals. ADCC mediated by KIR3DL1+ NK cells may represent a correlate of protection from HIV.

AIDS Vaccine 2011

53

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA02.05
Inhibition of Human Immunodeficiency Virus Type 1 Infection of Human Monocyte-Derived Macrophages by Anti-Lipid and Anti-MPER Monoclonal Antibodies
M. Rao1, O. Jobe2, K.K. Peachman2, G.R. Matyas1, L.V. Asher3, C.R. Alving1
1

Oral Abstract Session 02: Innate Immunity OA02.06 LB


JAK3 Inhibition In Vivo in Chronically SIV Infected Rhesus Macaques Deplete NK-17 Cells
L. Kowawisatsut1, A.A. Ansari2, K. Pattanapanyasat1 Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; 2Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
1

U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Rockville, MD, USA; 2U.S. Military HIV Research Program, Henry Jackson Foundation, Rockville, MD, USA; 3 Walter Reed Army Institute of Research, Silver Spring, MD, USA
Background: HIV-1 entry into cells requires the interaction of both HIV-1 envelope proteins and membrane lipids. We have generated several murine monoclonal antibodies (mAbs) that recognize the MPER region of HIV-1 envelope protein and/or lipids such as phosphatidylinositol-4-phosphate (PIP) and phosphatidylinositol4,5-bisphosphate. These lipids are present not only on the inner surface of plasma membranes of cells but also on the virions, which the virus acquires during the budding process. We investigated the ability of these mAbs to neutralize HIV-1 infection of primary monocyte-derived macrophages (MDM). Methods: Monocytes were isolated from peripheral blood mononuclear cells of HIV-1 seronegative donors and differentiated into MDM following in vitro culture. Binding of the mAbs to MDM was determined by flow cytometry and confocal microcopy. Detection of intracellular HIV-1 as well as murine mAb binding to US-1 virus was evaluated by electron microscopy. The neutralization ability of murine and human mAbs was assessed by flow cytometry. HIV-1 p24 and chemokines in the supernatants harvested from HIV-1 infected MDM cultures were assayed by ELISA. Results: Following preincubation with MDM, only WR301 (antiPIP IgM mAb) bound to the cells and effectively neutralized HIV-1. In contrast, the murine IgG mAbs WR324 (anti-MPER IgG mAb) and WR321 (anti-MPER, anti-PIP IgG mAb) effectively neutralized HIV-1 following preincubation with US-1. Electron microscopy showed that WR324 bound to virions. Infection of MDM enhanced the secretion of the chemokines several fold compared to MDM preincubated with mAbs alone. The chemokine levels were further increased in the presence of US-1 and the murine mAbs, with WR301 being a stronger inducer compared to WR324. Conclusion: The mAbs neutralize HIV-1 infection of MDM by different mechanisms. Antibodies generated against the lipid or against the HIV-1 envelope protein neutralize HIV-1 infection of primary human MDM either by production of chemokines or by binding to the virus.

Background: During simian immunodeficiency virus (SIV) infection, the gut-associated lymphoid tissue (GALT) is the primary target of virus replication during acute infection. While NK cells and IL-17 producing NK cells (NK-17) have been shown to play a key role in not only maintaining epithelial cell integrity of the GALT but also causing cytotoxicity for SIV infected cells. Their role during chronic infection is not clear. The purpose of this study was to determine the effect of NK cell depletion using a JAK3 inhibitor in chronically SIV infected rhesus macaques. Methods: Six chronic SIV-infected rhesus macaques were daily orally administered a pre-determined optimal dose of 10 mg/kg of a JAK3 inhibitor for 5 weeks. Blood and colorectal biopsies were collected. Cells were stimulated with PMA and ionomycin in the presence of brefeldin-A. The cells were stained with various fluorescent conjugated monoclonal antibodies and analyzed on LSRII flow cytometer. The plasma viral loads were also monitored. Results: Total NK cell and NK-17 in blood and biopsies were markedly depleted as early as the first week and the depletion persisted during the course of drug administration. While the total NK cells in blood returned to normal levels within 3 weeks after treatment cessation, the total NK cells in biopsies did not. The NK-17 cells in the blood and biopsies also did not return to normal levels after cessation. Drug treatment led to a transient increase in plasma viremia during the loss of total NK cells. Conclusion: The administration of a JAK3 inhibitor to chronically SIV infected monkeys depletes total and NK-17 in both the blood and mucosal tissue and is associated with increases in viremia denoting an important role for NK cells during chronic SIV infection.

ORAL ABSTRACT SESSIONS

54

AIDS Vaccine 2011

Oral Abstract Session 03: Novel Immunogens and Inserts OA03.01


Targeting Antigen to CD40 on Dendritic Cells Mediates Expansion of Multi-Epitope HIV-Specific Polyfunctional CD4+ and CD8+ T Cells
A. Flamar , Y. Xue , S. Zurawski , S. Oh , M. Montes , A. Montes , B. King2, L. Sloan2, J. Banchereau1, Y. Levy3, G. Zurawski1
1 1 1 1 1 1 1

Oral Abstract Sessions

OA03.02
Identification of Escape-Refractory Subdominant CD8 Epitopes for Common HLA Alleles
C.L. Boutwell1, C. Oniangue-Ndza1, A. Schneidewind1, H. Streeck1, A. Seese1, E. Mellors1, K. Murthy1, K. Power1, M. Kemper1, T. Dudek1, B.D. Walker1, M. Altfeld1, T.M. Allen1
1

Baylor Institute for Immunology Research, INSERM U899, Dallas, TX, USA; 2North Texas Infectious Diseases, dallas, TX, USA; 3UMR-S 955, HIV vaccine network/Vaccine Research Institute, Creteil, France
Background: Targeting Dendritic Cells (DCs) with anti-DC receptor antibody-antigen fusion proteins is a novel approach to vaccine development. In mouse models, these innovative vaccines induce potent cellular immune responses. Methods: We engineered an agonistic anti-human CD40 recombinant antibody fused via the heavy chain C-terminus to a string of five 19- to 32-amino-acid long sequences from HIV-1 Gag, Nef and Pol proteins (anti-CD40.HIV5pep). These peptides bear multiple highly conserved CD4+ and CD8+ T cell epitopes. HIV patient PBMCs or DC-T cell co-cultures were incubated with anti-CD40 and control hIgG4.HIV5pep fusion proteins. After 10 days, the total T cells were challenged with each individual HIV peptide, and then antigen-specific cytokine production was detected using intracellular staining. The cytotoxic function of the in-vitro expanded CD8+ T cell was also assayed. Results: Low doses of anti-CD40.HIV5pep prototype vaccine, but not the control hIgG4.HIV5pep, expand of peptide-specific CD4+ and CD8+ T cells. The range of antigen-specific T cells recall responses over the 5 HIV peptide regions of our antiCD40.HIV5pep varied between patients (n=9, range 1-4), but in sum epitopes from all 5 regions could be effectively processed and presented across the entire HIV-infected population we surveyed, which was heterogeneous for MHC alleles. These in vitro-expanded antigen-specific CD4+ and CD8+ T cells were polyfunctional, simultaneously producing multiple cytokines (IFNa, TNFa and MIP-1b), and the CD8+ T cells also had cytotoxic characteristics (granzyme B, surface CD107a, perforin). Finally the anti-CD40.HIV5pep-expanded CD8+ T cells were able to kill peptide-loaded autologous target cells and inhibit HIV-1 replication in vitro in autologous CD4+ T cells. Conclusion: Taken together, our results demonstrated that all desirable T cell effector qualities were elicited by our anti-CD40. HIV5pep prototype vaccine in the HIV-patient PBMCs and DC/T cell co-cultures. These in vitro data provide a pre-clinical rationale for further testing this DC-targeting HIV prototype vaccine for therapeutic applications in humans.

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA

Background: The mechanism of control of HIV by protective HLA class I alleles remains incompletely defined and thus so too does the immune environment to be induced by an effective HIV T cell vaccine. Here, we quantified the replicative cost of CTL escape in epitopes across the HIV proteome to explore the relationship between the differential cost of escape from immunodominant CD8+ T cell responses and the differential control of HIV by HLA alleles. Methods: We constructed recombinant HIV NL4-3 variants expressing each of 44 described HLA-associated polymorphisms in Gag and Nef located within 30 epitopes restricted by 16 HLA alleles. Replication capacity was quantified by a combination of FACS analysis in GFP-reporter cells and qRT-PCR in PBMC. Results: The majority of mutations did not significantly impact replication capacity, including escape mutations in the immunodominant epitopes of the non-protective HLA-A02 and A03 alleles. In contrast, 19 mutations significantly impacted viral replication, including the Gag mutations A163G, R264K, and K302R which reduced replication capacity by upwards of 60%. Notably, each of these mutations confers escape from immunodominant CD8 responses of the protective HLA-B57, B27, and B14 alleles. More importantly, Gag mutations K331R and E260D, located within epitopes targeted by sub-dominant HLA-B08 and B35 responses, also significantly impaired replication, revealing potentially durable responses lower in the targeting hierarchy of these common but non-protective HLA alleles. Conclusion: These data suggest that protective HLA alleles dominantly target epitopes in which escape occurs at high cost to the virus whereas non-protective alleles dominantly target epitopes in which escape occurs at minimal cost. The significant escape-associated costs observed in sub-dominant epitopes restricted by non-protective alleles suggest that vaccine antigens that induce responses against such escape refractory subdominant epitopes and avoid inducing responses against easily escaping immunodominant decoy epitopes may be central to the rational design of an effective HIV vaccine.

AIDS Vaccine 2011

55

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA03.03
HIV-1 gp120 V1V2-Scaffolds for Structural Analysis and Immunogen Design
J. McLellan1, M. Pancera1, K. Dai1, J. Boyington1, Z. Yang1, C. Carrico2, S. Shahzad-ul-Hussan1, M. Sastry1, S. Schmidt1, Y. Yang1, M. Bonsignori3, B. Haynes3, S. Phogat4, C. Bewley5, J. Mascola1, W. Schief2, G. Nabel1, P. Kwong1
1

Oral Abstract Session 03: Novel Immunogens and Inserts OA03.04


Identification of Mimotopes for Neutralizing Antibodies from Elite Controllers with EnvTailored Phage Display Libraries
M. Zhou1, S. Koch2, T. Meyer3, B. Brill1, M. Sakarellos-Daitsiotis4, J. Miguel Benito5, V. Soriano5, A. Haberl6, M. Bickel6, H. von Briesen2, M. Hust3, U. Dietrich1
1 Georg-Speyer-Haus Institute for Biomedical Research, Frankfurt, Germany; 2Global HIV Vaccine Research Cryorepository, Sulzbach, Germany; 3Technische Universitt Braunschweig, Braunschweig, Germany; 4University of Ioannina, Greece; 5Instituto de Salud Carlos III, Madrid, Spain; 6 University Hospital, Goethe University, Frankfurt, Germany

Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA; 2Department of Biochemistry, University of Washington, Seattle, WA, USA; 3Duke Human Vaccine Institute and the Duke Center for AIDS Research, Durham, NC, USA; 4International AIDS Vaccine Initiative, Brooklyn, NY, USA; 5 NIDDK, National Institute of Health, Bethesda, MD, USA
Background: To date, the only piece of gp120 that has not been determined at the atomic level is the V1V2-loop. Interestingly, portions of the V1, V2, and V3 loops are the target of broadly and potently neutralizing quaternary structure-preferring antibodies, such as the PG9, PG16 and CH01-05 antibodies. Although these antibodies prefer a quaternary epitope formed only in the context of the trimeric viral spike, they can bind to certain strains of gp120 monomers, albeit with reduced affinity. Methods: To obtain structural information on the V1V2-loop and understand how quaternary structure-preferring antibodies broadly neutralize diverse HIV-1 isolates whilst targeting loops that vary in sequence length and composition, we initiated structural and biophysical studies. The V1V2-loop from the primary isolate YU2 was transplanted into 17 acceptor scaffolds, and antigenic analysis was performed using monoclonal antibodies directed against the V1V2-loop of YU2. Results: Here we show that of the 17 YU2-V1V2 scaffolds expressed, two retained antigenic properties similar to V1V2 in full-length gp120. These two scaffolds are being used for structural studies by NMR and X-ray crystallography. A panel of gp120 was tested for binding to the quaternary structurepreferring antibodies, and the V1V2-loops from strains that showed binding were transplanted into the two previously characterized scaffolds. Conclusion: Structures of V1V2-loop scaffolds alone and in complex with neutralizing antibodies should aid in the design of immunogens capable of eliciting quaternary structurepreferring antibodies.

Background: The aim of this study was to identify immunogenic epitope mimics for HIV-neutralizing antibodies present in HIV1 elite controllers (EC), which are able to elicit neutralizing antibodies upon vaccination. Methods: Plasma samples from EC were tested for neutralization on TZM-bl cells against a cross-clade HIV pseudovirus panel. An Env-tailored phage display library was constructed and screened with serum antibodies from EC with cross-clade neutralizing capacity. The selected phage clones were analyzed by phageELISA for the binding specificity and peptide encoding inserts were sequenced. Phages and the corresponding linear peptides were tested for depletion of plasma neutralizing activity. The antibodies recognizing the selected epitopes were affinity purified from the corresponding serum and tested for neutralization. Mice were primed with pSyngp140JR-FL DNA and boosted with the selected epitope coupled to a palmitoyl, a sequential oligo peptide carrier (SOC) or to the phage. Results: A subgroup of EC was identified with cross-clade plasma neutralizing activity against pseudoviruses. An HIV-1 Envtailored phage library expressing random HIV-1 Env fragments was generated and screened with antibodies from EC26 who ranked top for neutralizing capacity. After several rounds of positive and negative selections the selected epitopes were sequenced. Epitopes were identified within the immunodominant regions of gp41, the fusion peptide and the membrane-proximal external region (MPER) of gp41. The MPER specific epitope EC26-2A4 could deplete approximately 50% plasma neutralizing activity against SF162.LS. Further, the affinity purified antibodies recognizing the EC26-2A4 epitope neutralized SF162.LS with an IC50 of 5.9 g/ml. Together these data prove the EC26-2A4 epitope as interesting vaccine candidate . The in vivo immunogenicity analysis of the EC26-2A4 epitope is ongoing. Conclusion: We demonstrated cross-clade neutralizing antibodies in a subgroup of EC and identified their antibodies based on Envtailored phage display libraries. Such epitopes may be suitable for the derivation of prophylactic vaccine candidates.

ORAL ABSTRACT SESSIONS

56

AIDS Vaccine 2011

Oral Abstract Session 03: Novel Immunogens and Inserts OA03.05 LB


Role of Hypervariable Loops in Intersubunit Associations of Env: Trimeric gp140 Immunogen as a Platform for Novel Epitope Display
H. Cheng1, C.G. Moscoso1, L. Xing1, L. Martin2, S. Barnett3, I. Srivastava4
1

Oral Abstract Sessions

OA03.06
Development and Characterization of HIV1 Virus-like Particles Produced by Stably Transfected Drosophila S2 cells
L. Yang1, Y. Song1, P. Zhu2, P. Zhou1
1 Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China; 2Institute of Biophysics, Chinese Academy of Sciences, China

University of California Davis, Davis, CA, USA; 2CEA, Gifsur-Yvette, France; 3Novartis, Inc., Cambridge, MA, USA; 4 National Institutes of Health, Gaithersburg, MD, USA
Background: The HIV-1 Env complex mediates membrane fusion with host cells following interaction with both a receptor and a coreceptor. The Env subunit gp120 binds to CD4 receptors, resulting in cryptic epitope exposure and allowing coreceptor binding (see our recent structure of Env gp140 trimer, Moscoso et al., 2011, PNAS 108(15):6091-6096). Some hypervariable loops on gp120, primarily V2 and V3, seem to play roles in the conformational transition to the induced, exposed coreceptor state. Methods: Here, we perform further comparative analysis of variants of the soluble Env construct gp140, with and without a partial V2 truncation, using cryoelectron microscopy and single particle reconstruction. Results: The Env structure displays a similar overall architecture with crucial detailed distinctions, one of them being a tighter association between gp120 subunits that results in a smaller trimer radius. Another pertinent feature is the presence of V2 near the base of the trimer proximal to the viral membrane, as suggested previously, placing V2 in proximity to the adjacent counterclockwise gp120 subunit. The juxtaposition of V2 and the neighboring gp120 subunits V3 loop (V3) contribute to a quaternary epitope formed by these two loops, which is the binding site for antibodies such as PG16 and 2909. Such a quaternary location of V2 would facilitate gp41-independent gp120-gp120 interactions, and hints at a mechanism of cooperative epitope occlusion mediated by hypervariable loop interaction, a decrease in trimer diameter and increased steric hindrance near the CD4 binding sites due to the decreased distance between adjoining CD4 epitopes. Conclusion: Comparative structural analysis of these recombinant vaccine candidates informs rational design of strategies towards an immunogen with conserved structural features based on the native form of Env trimers, previously missed in monomeric gp120 vaccine efforts, to maximize exposure of novel epitopes and thus to elicit broadly and potently neutralizing antibodies, such as VRC01.

Background: HIV-1 virus-like particles (VLP), perceived to be much safer than live-attenuated viruses, are being developed as a vaccine candidate against HIV-1/AIDS. However, in one way or the other current methods used for HIV-1 VLP production have many limitations, such as low yield, poor gp160 cleavage, difficulty to purify and variation between batches. Methods: To overcome these limitations, we developed a Drosophila S2 expression system for HIV-1 VLP production and evaluated immunogenicity of S2-produced HIV-1 VLP in heterologous DNA-VLP prime-boost strategy in mice. Results: Here we report that stable S2 cell transfectants efficiently produce and release HIV-1 VLP into culture supernatants with the amount much higher than those produced by transiently transfected 293 T cells. HIV-1 envelope proteins are properly processed and glycosylated in a high mannose form. HIV-1 envelope and gag proteins both are effectively incorporated into VLP. Cryo electron microscopy indicates that HIV-1 VLP contains many HIV-1 spikes on VLP surface. Finally, we show that heterologous DNA-VLP prime-boost elicits both ELISA-binding and neutralizing antibody responses as well as HIV-1 envelope and gag peptide-specific CD8 and CD4 T cell responses.

AIDS Vaccine 2011

57

ORAL ABSTRACT SESSIONS

Conclusion: Thus, we conclude that production of HIV-1 VLP by stable transfectants of Drosophila S2 cells overcomes several limitations of the current HIV-1 VLP producing systems. The HIV1 VLP produced by stable S2 transfectants elicits both hormoral and cellular immune responses.

Oral Abstract Sessions


OA04.01

Oral Abstract Session 04: Novel Monoclonals and Structural Insights OA04.02
Human Anti-V2 Monoclonal Antibodies Neutralize Tier 1 HIV-1 Pseudoviruses
M.K. Gorny1, C. Williams1, T. ONeal1, X. Wang2, M.S. Seaman3, S. Zolla-Pazner1 New York University School of Medicine, New York, NY, USA; 2Veterans Affairs New York Harbor Healthcare System, New York, NY, USA; 3Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
1

Potent and Broad Neutralization by a CD4 Binding Site Monoclonal Antibody from an HIV-1 Infected Donor
E. Falkowska , D.R. Burton , P. Poignard , J. Mascola , P.D. Kwong3, X. Wu3
1 2 1 3 1

International AIDS Vaccine Initiative and Neutralizing Antibody Center at The Scripps Research Institute, La Jolla, CA, USA; 2International AIDS Vaccine Initiative/Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 3 Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
Background: Recently, several novel neutralizing antibodies (nAbs) have been isolated from HIV-1 positive donors. Of these nAbs, PG9/16, and VRC01, are unprecedented in their potency and breadth. In this study, we characterize PGV04 (also known as VRC-PG04), a new nAb that has potency and breadth that rivals that of PG9/16 and VRC01. Methods: PGV04 was isolated by single, memory B cell sorting using the resurfaced core (RSC3) protein as bait. The nAb was screened for neutralization on a 162 virus panel and shown to be both broad and potent. The epitope was further mapped by competition with other MAbs and by evaluating binding and neutralization of single alanine substitutions in the JR-CSF gp120 protein monomer and gp120 incorporated into pseudovirus respectively. Binding to cell surface expressed trimers was measured by flow cytometry and binding to deglycosylated gp120 was measured by ELISA. Results: PGV04 competed with CD4, b12 and VRC01 for binding to gp120, confirming it is a CD4bs mAb. However, when screened on a large panel of viruses, PGV04 varied in its neutralization profile from VRC01, VRC03 and b12. Furthermore, PGV04 binding to gp120 containing single alanine substitutions revealed differences in residue dependence between PGV04 and other CD4bs mAbs. The residues found to be important in binding to gp120 had varying effects on the ability of PGV04 to neutralize pseudovirus incorporating these same alanine substitutions and only one substitution, D279A, abrogated PGV04 neutralization completely. All of the CD4bs mAbs tested bound deglycosylated gp120 and none induced the co-receptor site on functional trimers. Conclusion: Broad and potent CD4bs nAbs have subtle differences in the way they recognize and access the CD4bs in the context of the viral spike.

Background: Antibodies (Abs) to the V2 region of gp120 are present in a minority of HIV-infected subjects, and, to date, little has been published on their immunologic or functional properties. Recent analysis of plasma from the RV144 clinical vaccine trial, however, revealed that anti-V2 Abs were present in almost all plasma from vaccine recipients, and their contribution to protection is being extensively analyzed. Methods: A panel of seven human anti-V2 monoclonal Abs (mAbs), generated previously using cellular techniques from PBMCs of individuals infected with clade B HIV-1, was analyzed for their immunoglobulin variable gene usage, ELISA crossreactivity to recombinant gp120 derived from viruses from clades A, B and C, and neutralizing activity against pseudoviruses using the TZM-bl cell assay. Results: All the anti-V2 mAbs are IgG1 except for one which is IgG3. Five mAbs were sequenced: four use the same IGHV169 gene segment, suggesting preferential gene usage by Abs targeting this region. The remaining mAb uses the IGHV434 gene and has very different immunologic characteristics, including interference with sCD4 binding. All seven V2 mAbs are highly cross-reactive with comparable relative affinities. They bind to 19 recombinant gp120s representing eight from clade B, ten from clade C and, one from clade A. Four anti-V2 mAbs were tested for their neutralizing activity. Tier 1 pseudoviruses were neutralized, including six from clade B and two from clades A and C. The 50% neutralizing values ranged between <0.8 and 88.7 g/ml, with a mean of 16.6 g/ml. Conclusion: The results indicate that human anti-V2 mAbs display cross-clade neutralization against Tier 1 pseudoviruses in the TZM.bl assay.

ORAL ABSTRACT SESSIONS

58

AIDS Vaccine 2011

Oral Abstract Session 04: Novel Monoclonals and Structural Insights OA04.03
2F5-like Cross-Reactive HIV-1-Neutralizing Monoclonal Antibodies with Low Number of Somatic Mutations
Z. Zhu5, H.R. Qin5, W. Chen5, Q. Zhao5, X. Shen1, R. Schutte2, Y. Wang5, G. Ofek3, E. Streaker5, P. Prabakaran5, G.G. Fouda2, H. Liao2, J. Owens5, M. Louder3, Y. Yang3, K. Klaric4, M.A. Moody2, J.R. Mascola3, J.K. Scott4, P.D. Kwong3, D. Montefiori2, B.F. Haynes2, G.D. Tomaras2, D.S. Dimitrov5
1

Oral Abstract Sessions

OA04.04
Characterization of CD4-Binding Site Directed Monoclonal Antibodies Isolated from HIV-1 gp140 Env Immunized Rhesus Macaques
C. Sundling1, Y. Li2, N. Huynh1, R. Wilson2, S. ODell3, J.R. Mascola3, R.T. Wyatt2, G.B. Karlsson Hedestam1 Karolinska Institutet, Stockholm, Sweden; 2Department of Immunology and Microbial Science, The Scripps, La Jolla, CA, USA; 3Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
1

Vaccine Institute, Durham, NC, USA; 2Human Vaccine Institute, Durham, NC, USA; 3Vaccine Research Center, Bethesda, MD, USA; 4Simon Fraser University, Burnaby, BC, Canada; 5NCI-Frederick, NIH, Frederick, MD, USA
Background: The genes encoding broadly HIV-1-neutralizing (bn) human monoclonal (m) antibodies (Abs) are highly divergent from their germline counterparts. We have hypothesized that this could pose a challenge for their elicitation and that identification and characterization of less somatically mutated bnAbs may help in the design of effective vaccines Methods: IgG and IgM libraries were constructed using PBMC mRNA from a patient with bn serum containing Abs targeting the epitope of the bn mAb 2F5. The mAbs were selected from the libraries by panning against peptides containing the 2F5 epitope and against HIV-1 gp140JR-FL, and characterized by standard assays. Results: Two mAbs (m66 and m66.6) were identified that share the same heavy chain; the variant bearing the more mutated light chain (m66.6) exhibited higher HIV-1 neutralizing activity than the less mutated variant - it was very potent neutralizer in assays based on PBMCs and macrophages but less potent than 2F5 in the TZM-bl assay. Both mAbs required the DKW664-666 core of the 2F5 epitope as well as two additional upstream residues (L660,663) for binding. The heavy chains of these mAbs have long (21 residues) third complementarity determining regions. m66.6 exhibited relatively weak polyspecific reactivity to a few self antigens and m66 was not autoreactive. Both m66 and m66.6 use different VH and VL genes from 2F5 and are significantly less divergent from their germline Ab counterparts than 2F5 they have a total of 11 and 18 amino acid changes, respectively, from the closest VH and V germline gene products, compared to 25 for 2F5 Conclusion: m66.6 and m66 represent unique Abs whose binding mechanism and neutralization specificities overlap with those of 2F5. This pair of mAbs should be of use in exploring the complex maturation pathways of bnAbs, poly- and autoreactivity, and may inform the design of vaccine immunogens and HIV therapeutics.

Background: An increased focus on the cells that produce vaccine-elicited antibodies (Abs) and on the process of B cell maturation can help guide efforts aimed at replicating the successful generation of broadly neutralizing Abs observed in some infected humans. Examination of neutralizing monoclonal Abs (MAbs) from chronically infected individuals suggests that extensive Ab affinity maturation is required to achieve efficient neutralization; however, little is known about the relationship between HIV-1 protein subunit immunization and Ab somatic hypermutation. Methods: We have isolated and expressed Abs from single cellsorted Env-specific memory B cells from non-human primates (NHPs) that were immunized with soluble YU2 gp140 trimers. The sorting was performed with Env probes designed to selectively isolate CD4 binding site (CD4bs)-specific B cells. PCR amplification of Ab heavy and light chain gene transcripts was followed by sequence analysis, expression and characterization of MAbs. Results: Analyses of a first panel of eight MAbs demonstrates that all eight arose from different clonal lineages as determined by their VDJ gene family usage. All MAbs were capable of neutralizing Tier 1 strains of HIV-1 with reasonable potency, including some non-clade B viruses. Their neutralization pattern was consistent with the polyclonal plasma activity elicited in the animal from which the MAbs were isolated. Cross-competition analyses demonstrated that all eight NHP MAbs cross-competed with a set of well-characterized CD4bs-directed human MAbs, confirming their specificity for the CD4bs consistent with the criteria used for NHP memory B cell sorting. Several of the MAbs displayed distinct binding properties when tested against a panel of gp120-derived ligands. Conclusion: These data show that the soluble gp140 trimers elicit neutralizing CD4bs-directed Abs in healthy non-human primates. Further examination of the fine specificity of these and additional MAbs will provide information to guide the design of improved vaccines and immunization strategies.

AIDS Vaccine 2011

59

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA04.05
Structural Insights into HIV-1 Neutralization by New Highly Potent and Broadly Neutralizing Antibodies
J. Julien3, R. Pejchal3, L. Kong3, R.S. Stanfield3, J. Lai3, L.M. Walker3, K.J. Doores1, M. Huber1, E. Falkowska1, P. Poignard3, S.K. Phogat2, W.C. Koff3, D.R. Burton1, I.A. Wilson3
1 The Scripps Research Institute and Ragon Institute of MIT, La Jolla, CA and Boston, MA, USA; 2The International AIDS Vaccine Initiative, New York, NY, USA; 3The Scripps Research Institute, La Jolla, CA, USA

Oral Abstract Session 04: Novel Monoclonals and Structural Insights OA04.06
AAV2/8-Encoded Vaccine Expressing a Broadly Neutralizing HIV Antibody Protects Humanized Mice from High-Dose Intravenous Viral Challenge
A.B. Balazs1 , J. Chen1, C.M. Hong1, D.S. Rao2, L.Yang1, D. Baltimore1 Division of Biology, California Institute of Technology, Pasadena, CA, USA; 2Deptartment of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
1

Background: Human monoclonal antibodies have been characterized recently that potently neutralize HIV-1 isolates across all clades. These exciting new antibodies (PGT series) were derived from direct functional screening of B cells from IAVI protocol G donors (Theraclone/Monogram) and are unusually potent with binding predicted to be to novel epitopes on Env gp120. Methods: The crystal structures of these new PGT antibodies are being determined by x-ray crystallography with further characterization using binding and mutagenesis data. Results: The crystal structures so far have been elucidated for one complete family of these antibodies and others are in progress, as well as Fab complexes with gp120 and fragments. Conclusion: Structural characterization and biochemical analysis of these antibodies have uncovered novel specificities to new epitopes and reveal further mechanisms for viral neutralization. These new epitopes provide additional insights for neutralization of HIV-1 and can be used for structure-assisted vaccine design.

Background: Despite spending nearly a billion dollars globally every year on HIV/AIDS research, over 25 million people have died as a result of the ongoing pandemic. In spite of the failure of the humoral response to protect individuals from HIV infection, several monoclonal antibodies such as b12, 2F5, 4E10, and 2G12 have been identified that are capable of neutralizing a broad range of primary HIV isolates. Because of this, significant effort has been focused on the design of immunogens capable of eliciting antibodies de-novo that would employ similar modes of recognition and target similar epitopes as these clones. As an alternative strategy, we have designed a novel geneticallyencoded vaccine in which the expression of specific monoclonal antibodies is directed by Adeno-Associated Virus engineered to produce Human IgG1. Methods: Following a single intramuscular injection of this vaccine, mice produced nearly 1mg/mL of circulating human b12 antibody from a single 1011 dose. To test the efficacy of this vaccine we humanized vaccinated mice by engrafting Human peripheral blood mononuclear cells into immunocompromised NOD/SCID/-/animals prior to challenge with a pathogenic HIV strain (NL4-3). Results: Vaccinated mice exhibited remarkable resistance to both peripheral and splenic CD4 loss following challenge. Using this system we have quantified the protective abilities of four well-characterized broadly neutralizing antibodies in vivo. Interestingly, animals protected by the b12 neutralizing antibody appear to have undetectable levels of infection as measured by histological analysis of the spleen, suggesting that sterilizing immunity may be the principal mechanism of protection. Consistent with this result, animals challenged with escalating doses of HIV demonstrated robust protection of CD4+ T-lymphocytes despite artificially high levels of inoculum. Conclusion: Future experiments aim to utilize this model to establish minimum protective doses of neutralizing antibodies and define concentration targets for human clinical trials.

ORAL ABSTRACT SESSIONS

This study was supported by the International AIDS Vaccine Initiative (IAVI), Ragon Institute, and NIH AI84817.

60

AIDS Vaccine 2011

Oral Abstract Session 04: Novel Monoclonals and Structural Insights OA04.07 LB
Evolution of HIV-1 Transmitted/Founder Viruses Results in the Formation of Epitopes for Later Broadly Cross-Neutralizing Antibodies
P.L. Moore1, E.S. Gray1, T. Hermanus1, D. Sheward2, N. Tumba1, C.K. Wibmer1, J. Bhiman1, S. Sibeko3, S.S. Abdool Karim3, C. Williamson2, L. Morris1
1 AIDS Virus Research Unit, NICD, South Africa, Johannesburg, South Africa; 2University of Cape Town, Cape Town, South Africa; 3CAPRISA, University of KwaZulu Natal, Durban, South Africa

Oral Abstract Sessions

Background: Much emphasis has been placed on mapping the epitopes of broadly cross-neutralizing (BCN) antibodies. However, the interplay between strain-specific antibodies, BCN antibodies and autologous viral evolution has not been well characterized. Methods: Single genome amplification was used to obtain longitudinal autologous envelope sequences from three individuals (CAP177, CAP8 and CAP256) in the CAPRISA 002 cohort who developed BCN antibodies with known specificities (Gray et al, JV, 2011 and Moore et al, JV, 2011). The sequences were then analysed to determinewhether previously defined BCN epitopes were present on the transmitted/founder viruses, and to characterize their evolution over time. Results: CAP177 developed BCN antibodies dependent on a glycan at position 332 in the C3 region. This N332 glycan was however not present on the CAP177 transmitted/founder virus, but arose by 6 months post-infection as a result of escape from an early strain-specific antibody targeting the alpha-2 helix of the C3 region. Similarly in CAP8, who developed BCN antibodies dependent on the N160 glycan, the transmitted founder virus did not encode the N160 glycan, but instead contained a K160 residue. The N160 glycan evolved in CAP8 by 6 months of infection and became fixed. In a third individual, CAP256, who also developed extremely potent N160-dependent BCN antibodies, the transmitted/founder virus lacked this residue. However, superinfection at 15 weeks post-infection with a second virus that contained the N160 glycan resulted in the generation of recombinant viruses that harbored the N160 glycan forming the CAP256 BCN epitope. Conclusion: In three individuals who developed BCN antibodies, the BCN epitope was not present on the transmitted/founder virus, but evolved within 6 months of infection. In one case, the BCN epitope evolved via viral escape from an earlier strain-specific antibody targeting the same region. These studies highlight the role of viral evolution in shaping the development of BCN antibody responses.

AIDS Vaccine 2011

61

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA05.01
HIV-Specific Cytolytic CD4+ T-Cell Responses During Acute HIV-1 Infection Predict Disease Outcome
D.Z. Soghoian1, H. Jessen2, S. Cutler1, M. Flanders1, S. Ranasinghe1, M. Lindqvist1, I. Davis1, K. Lane1, J. Rychert3, E.S. Rosenberg3, B.D. Walker1, H. Streeck1
1

Oral Abstract Session 05: Acute Infection/Viral Diversity OA05.02


Control of Acute HIV-1 Subtype C Viremia Is Associated with Limited CD8+ T-Cell Immunogenicity
M.A. Radebe1, K. Nair1, F. Chonco1, K. Bishop1, K. Bishop1, J.K. Wright1, M. van der Stok1, Z. Mncube1, M. Altfeld2, B.D. Walker2, T. Ndungu1
1

Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA, USA; 2Praxis Jessen Jessen Stein, Berlin, Germany; 3 Infectious Disease Division, Massachusetts General Hospital, Boston, MA, USA
Background: During acute HIV-1 infection, early control of viremia and establishment of viral set point have been widely attributed to HIV-specific CD8 T-cell responses. However, despite increasing evidence for direct antiviral activity by CD4 T-cells in other infections, the impact of HIV-specific CD4 T-cells on viral control has not been studied. Methods: We studied 26 acutely infected, treatment-nave subjects and monitored their clinical outcome for up to 3 years post-infection. Baseline HIV-specific CD4 T-cell responses were assessed for degranulation (CD107a), IFN secretion, expression of granzymes (GrzA,B,K) and perforin. HIV-specific cytolytic CD4 T-cell responses were investigated longitudinally in a subset of 11 subjects with similar peak viral loads for 1 year post-infection. Results: Among the patients followed longitudinally, 6 progressed to a low viral set point, while 5 progressed to a significantly higher set point (134,020vs.11,234 copies/ml; p=0.004). Interestingly, a significant expansion of HIV-specific cytolytic CD4 cell responses was observed in individuals who controlled viral replication compared to those who progressed to a high viral set point (IFN: p=0.038, CD107a: p=0.042). Importantly, this expansion was observed early post-infection, prior to the divergence of viral load or CD4 T cell counts between the two groups. Examination of the baseline HIV-specific CD4 responses revealed a distinct GrzAenriched cytolytic phenotype that was highly associated with subsequent viral control. Strikingly, Kaplan-Meier analysis of only the baseline cytolytic CD4 T-cell response in a larger cohort demonstrated that individuals with HIV-specific GrzA+ responses remained off HAART significantly longer than individuals with HIV-specific GrzA- responses (p=0.0023). Conclusion: Here we demonstrate that the rapid induction and expansion of a distinct cytolytic CD4 T-cell response during acute infection is significantly associated with viral control and disease outcome. These data suggest a pivotal role for HIV-specific cytolytic CD4 T-cells in the early control of viremia following acute infection and for future HIV vaccine design strategies.

University of KwaZulu-Natal, Durban, South Africa; 2Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Boston, MA, USA

Background: HIV-1-specific CD8+ T-cell responses contribute to the decline in acute peak viremia following infection. However, the relative immunogenicity CD8+ T-cell epitopes during and after acute viremia has not been well defined, and data on the breadth of responses following the initial drop in acute viremia are lacking. Methods: We characterized CD8+ T-cell responses to peptides spanning the full viral proteome in 20 HIV-1C acutely infected, antiretroviral naive subjects using the IFN- ELISPOT assay as early as 28 days after estimated date of infection. Viruses from plasma were also sequenced within defined CD8+ T-cell epitopes for selected subjects. Of the 20 participants in the cohort, eleven had not reached complete seroconversion at analysis. Results: Despite documented declines in peak viremia, the initial responses associated with the decline in acute phase peak viremia were narrowly directed, and of weak magnitude. At approximately 28 days after estimated initial infection, following a 15-fold decline in viral loads, CD8+ T-cell responses were directed against an average of 3 of the 410 peptides tested (range 06); two individuals had no detectable CD8+ T-cell responses at this time. At 18 weeks post the estimated date of infection the average number of peptides targeted had increased to 5 (range 0-11). Of 56 optimal Gag CD8+ T-cell epitopes sequenced, 31 were wild type in the acute infecting viruses, however, only 11 of 31 elicited measurable CD8+ T-cell responses. Conclusion: These data demonstrate that the majority of CD8+ responses that subsequently arise are not elicited during acute HIV-1 infection despite the presence of the cognate epitope in the infecting strain. Further studies are needed to address why recognition of HIV-1 peptides appears to be selective during acute infection as the lack of recognition may contribute to the failure to control viremia to a low set point.

ORAL ABSTRACT SESSIONS

62

AIDS Vaccine 2011

Oral Abstract Session 05: Acute Infection/Viral Diversity OA05.03


Lymphocyte Dynamics in Acute HIV-1 Infection: RV217-The Early Capture HIV Cohort Study (ECHO)
L. Eller5, M. Eller5, A. Eser1, P. Naluyima2, K. Shikuku3, A. Taylor2, E. Rono3, S. Sukwit4, S. Nitayaphan4, J. Currier5, M. de Souza5, S. Peel5, J. Kim5, N. Michael5, M. Marovich5, M. Robb5
1 Mbeya Medical Research Program, Mbeya, United Republic of Tanzania; 2Makerere University Walter Reed Project, Kampala, Uganda; 3Walter Reed Project Kericho, Kericho, Kenya; 4Royal Thai Army, Department of Retrovirology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 5US Military HIV Research Program (MHRP), Rockville, MD, USA

Oral Abstract Sessions

OA05.04
Long-Lived Historical Record of Evolution of Viral Diversity in Peripheral Blood Cells by Deep Pyrosequencing
L. Yin1, L. Liu1, Y. Sun1, W. Hou1, A.C. Lowe1, M. Salemi1, W.G. Farmerie1, J.W. Sleasman2, M.M. Goodenow1
1

University of Florida, Gainesville, FL, USA; 2University of South Florida, St. Petersburg, FL, USA

Background: Assessments of HIV-1 diversity are generally inferred from limited population sampling. Deep sequencing was applied to increase quasispecies coverage for direct analysis of viral diversity, population structure, and the historical archive of HIV-1 within the host. Methods: 454 deep sequencing was applied to HIV-1 envelope hypervariable domain 3 (V3) in peripheral blood cells from therapy-nave children, infected for 1.5 to 6.1 years, with high plasma virus levels (4.0 to 6.7 log10 HIV-1 RNA copies/ml), but variable CD4 T-lymphocytes (2% to 30%). A V3 library was constructed for each subject from 400 viral templates and sequenced to generate 10,000 quality reads. Consensus sequences were derived by clustering reads at 3% genetic distance, and used to estimate viral biodiversity and population structure via rarefaction/CHAO1 algorithm. Coreceptor use was inferred by position-specific scoring matrix. Viral archive and most recent common ancestor were evaluated by maximum likelihood phylogenetic trees constructed from consensus sequences combined with longitudinal conventional sequences. Results: Biodiversity of HIV-1 V3 quasispecies across individuals ranged from 12 to 99 clusters. Viral population structure was organized into a limited number of dominant genomes with a high frequency of unique sequences. Dominant viral variants at later time points evolved exclusively from low frequency variants at earlier time points. Phenotypic profiles of bioclusters revealed a breadth of coreceptor use that was unrelated to population structure or immune status. CXCR4 variants were co-archived with CCR5 variants even when CD4 T-cells exceeded 20%. Viral variants persisted in peripheral blood cells for as long as six years. In at least one individual, pyrosequencing at four years after infection identified minor viral variants that colocalized with early transmitting viruses. Conclusion: Unprecedented coverage by deep sequencing defines HIV-1 population complexity and structure, enriches the evolutionary landscape between samplings, and reveals an evolutionary record of persistent, cell-associated viral sequences closely related to transmitting viruses.

Background: Acute HIV infection events between exposure and peak viremia in non-subtype B populations are poorly characterized. Methods: Individuals in East Africa and Thailand at high risk for HIV-1 infection were prospectively followed with twice weekly HIV-1 RNA (NAT) to detect acute infection. Lymphocyte subsets (CD4+T, CD8+T, NK and B cells) were enumerated on a FACSCalibur using MultiTest reagents (Becton Dickinson) starting as early as 3-9 days after the last known NAT-negative sample and followed at multiple timepoints during early infection. Viral load (VL) was measured using Abbott Real Time Viral Load. Results: Dynamic lymphocyte changes were observed in 16 acute HIV-1 infections. Peripheral CD4+T cell and B cell nadir occurred at a median of 20 days post infection. CD4+T cells fell from a median 929 (range, 419-1532) cells/ul to 466 (142-866) cells/ul (P<0.001). B cells declined from a median 290 (186592) cells/ul to 127 (15-275) cells/ul (P<0.001). In contrast, NK and CD+T cells increased at different rates within the first 100 days. CD8+T cells increased nearly 3-fold from a median 500 (271-1150) cells/ul to 1498 (693-2367) cells/ul (P<0.001). NK cells increased from a median 252 (49-589) cells/ul to 335 (100864) cells/ul (P=0.002). CD4+T cell and B cell counts exhibited an inverse correlation (P<0.001) while NK cell counts showed a direct correlation (P=0.022) with contemporaneous VL. CD8+T cell counts did not correlate with VL at any time consistent with expansion by indirect mechanisms. VLs preceding and following CD4+T cell and B cell measurements were negatively correlated suggesting that these subsets are being directly depleted. In contrast, VL preceding, but not following NK cell measurements were positively correlated, suggesting that VL may be driving NK cell expansion. Conclusion: Understanding lymphocyte dynamics, their relationship to VL and early pathogenic events of non-B HIV-1 infection may help to target prevention or treatment interventions.

AIDS Vaccine 2011

63

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA05.05
Dynamics of HIV-1 Subtype C Gag in the Global Epidemic
V. Novitsky1, R. Wang1, M. Essex1
1

Oral Abstract Session 05: Acute Infection/Viral Diversity OA05.06


Important Contribution of Subdominant HIVSpecific CD8 T Cell Responses to Early Control of HIV Replication
H. Streeck1, D. Kwon1, M. Addo1, M. Flanders1, I. Davis1, S. Cutler1, J. Rychert1, K. Lane1, B. Yassine-Diab1, J. Routy1, S. Little2, H. Jessen3, A. Kelleher1, F. Hecht4, R. Sekaly1, E. Rosenberg1, B. Walker1, T. Allen1, M. Altfeld1
1 Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2University of California, San Diego, San Diego, CA, USA; 3Practice Jessen, USA; 4University of California, San Francisco, San Franciscon, CA, USA

Harvard School of Public Health, Boston, MA, USA

Background: The dynamic patterns of HIV transmission, and the complexity of viral diversity on the population level, might present substantial challenges to HIV vaccine design. It is unclear to what extent HIV vaccine antigens should match viral evolution to provide a sufficient level of protection. Methods: To address HIV-1 subtype C evolution on a population level over time, 1,520 unique gag sequences were retrieved from the LANL HIV Database, grouped by sampling year into 9 sets (19831989, 19901998, 1999, 2000, 20012002, 2003, 2004, 20052006, and 20072008), and analyzed for site-specific frequency of translated amino acid residues and tMRCA. Results: Despite the heterogeneous origins of the analyzed sequences, the gamut and frequency of amino acid residues in wild-type Gag were remarkably stable over the past two decades of the HIV-1 subtype C epidemic. The vast majority of amino acid residues demonstrated minor frequency fluctuation over the observed time, consistent with the conservative nature of the HIV1 Gag protein. Consistent changes in amino acid frequency were detected in only 4.0% (20/500) of amino acid residues across Gag based on three statistical methods, a trend test, heterogeneity test, and range ranking of amino acid frequency. The tMRCA of HIV-1 subtype C was dated to around 1959 (95% HPD 19541964). Conclusion: The study presents evidence of the stability of HIV-1 subtype C Gag among viruses circulating in the epidemic over the past two decades, providing an optimistic prognosis for HIV antigens tested in vaccine trials. On a background of the overall increase of HIV-1 subtype C gag diversity over time, a relatively small number of amino acid residues across HIV-1 subtype C Gag are likely to be under consistent selection pressure at the population level. The year 1959 was estimated as the beginning of HIV-1 subtype C diversification in the worldwide epidemic.

Background: During acute HIV-1 infection, HIV-specific CD8 T cell (CTL) responses emerge, suppressing viral loads to a semistable viral setpoint; this early viral setpoint has been shown to be highly predictive of disease outcome. However, while immunodominant responses restricted by rare HLA class I alleles have been associated with better control of viral replication, immunodominant responses restricted by common HLA alleles like B*8 or B*7 do not appear to have a significant impact on early control. Methods: Over 600 individuals with primary HIV-1 infection were screened for HLA-restricted, epitope-specific CTL responses using optimally defined epitopes. The early viral setpoint was determined in all treatment-nave individuals by two independent ID specialists. Results: The recognition of HIV-specific CTL responses was very predictable based on HLA class I expression and followed a clear hierarchical pattern. While the immunodominant responses in individuals with rare HLA class I alleles were significantly associated with lower viral setpoint, individuals with immunodominant responses restricted by common HLA class I alleles had, on average, higher viral loads. Interestingly, the absence of CTL responses in primary infection directed against some of the immunodominant epitopes restricted by common HLA class I alleles (e.g.HLA-B8-FL8) allowed for the development of responses against subdominant epitopes (e.g.HLA-B8-EI8) within the same individuals and was associated with significantly better subsequent viral control(p=0.001). A distinct ranking of the relative contribution of epitope-specific CTL responses to the early viral setpoint suggested an unexpected contribution of subdominant responses to the initial viral control. Conclusion: Our data suggest that in the context of common HLA class I alleles, CTL responses directed against subdominant epitopes but not dominant epitopes during primary infection significantly contribute to better control of viremia. Thus, engineered proteins that lack common immunodominant CTL epitopes and allow for the priming of otherwise subdominant epitopes might provide an important approach for vaccine design strategies.

ORAL ABSTRACT SESSIONS

64

AIDS Vaccine 2011

Oral Abstract Session 06: T-Cell Immunity and Immune Escape OA06.01
Deconstructing HIV-Specific CD8+T Cell Responses in HIV Elite Controllers
Z.M. Ndhlovu1, L.B. Chibnik2, J. Proundfoot1, S. Vine1, F. Porichis1, A. McMullen1, K. Cesa1, A. Piechocka-Trocha1, P.L. De Jager2, D.E. Kaufmann1, B.D. Walker1
1

Oral Abstract Sessions

OA06.02
Ontogeny of CD8+ T Cells in Acute HIV-1 that Inhibit Autologous and Heterologous Transmitted/ Founder (T/F) Viruses
S.A. Freel1, R.A. Picking1, G. Ferrari1, C. Ochsenbauer2, J.C. Kappes2, J. Kirchherr1, K.J. Weinhold1, K. Soderberg1, A.J. McMichael3, B.F. Haynes1, G.D. Tomaras1
1 2

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2Brigham and Women Hospital, Harvard Medical School, Boston, MA, USA

Duke University Medical School, Durham, NC, USA; University of Alabama, Birmingham, AL, USA; 3University of Oxford, Oxford, United Kingdom (Great Britain)

Background: Evidence suggests that HIV-specific CD8+T cells (CTL) play a critical role in a large proportion of rare individuals capable of immune control of HIV replication in the absence of therapy (elite controllers, EC). However, there is still no clear understanding of what differentiates effective CTL responses from ineffective responses in untreated chronic progressive disease (CP) and in subjects with controlled viral load on antiviral therapy (ARTC). Methods: We combined standardized assays and advanced statistical analysis to determine whether a subset of HIV-specific CTL endowed with unique characteristics is present in EC. Inter and intra-donor analysis of HIV CTL-specific proliferation and cytokine secretion was performed at different time points after antigen stimulation by CFSE and luminex assays. 23 EC, 15 CP and 15 ARTC were studied. Independent cohorts of 15 EC and 15 ARTC were used for validation. Results: We observed that within the same individuals, the proliferative capacity of CTL targeting epitopes restricted by protective alleles was significantly stronger than epitopes restricted by non-protective HLAs (p =0.003). However, this marked difference was present only in EC. Kinetic analysis of cytokine secretion showed marked differences between groups. Slopes of IFN-, IL-2 and TNF secretion were significantly steeper in EC than in the other groups. A model incorporating proliferation and early IL-2 secretion accurately predicted EC status in an independent cohort (c-statistic=0.908) Conclusion: Our data illustrate that HIV-specific CTLs in EC are characterized by late and robust cytokine secretion. We also show that proliferation is driven by protective alleles only in EC, suggesting that HIV-specific responses restricted by protective HLA alleles present unique features in EC not observed in subjects with progressive disease. Our results suggest that this strategy can be used to define immune correlates of protection in natural HIV infection and may have important implications for immune monitoring of responses to HIV

Background: CD8-mediated virus inhibition can be detected in HIV-1+ subjects naturally controlling virus replication. Characterizing the inhibitory function of CD8+ T cells in acute HIV-1 infection (AHI) is important for determining the nature of CD8+ responses that need to be elicited by an HIV-1 vaccine. In this study, we examine the timing, epitope specificity, and contribution of soluble factors to the antiviral CD8+ T cell response in AHI. Methods: We used a CD8+ T cell mediated virus inhibition assay (CD8 VIA) to assess CD8+ T cell function during AHI in the CHAVI 001 cohort. Primary CD4+ enriched lymphocytes were infected with transmitted/founder (T/F) autologous and heterologous viruses in the presence of autologous CD8+ T cells. Peptide-specific soluble antiviral responses were determined by transwell assay, luminex cytokine analysis, and flow-cytometric intracellular cytokine staining. Results: Potent CD8+ antiviral responses against autologous and heterologous (T/F) viruses appeared during AHI prior to Fiebig stage 4. Responses against autologous transmitted virus were durable to 48 weeks; however heterologous responses declined concurrent with the resolution of viremia. Viruses obtained 6 months post infection were more resistant to CD8+ mediated virus inhibition than cognate T/F viruses. CD8+ T cells specific for epitopes that have been shown to drive HIV-1 escape in AHI released soluble factors that inhibited T/F virus replication. Conclusion: The rapid development of potent CD8+ antiviral activity may reflect the previously identified inhibitory capacity of early and transitional memory cells. These studies suggest that continued antigenic exposure is necessary to maintain antiviral breadth. Chronic virus resistance to early CD8+ cell antiviral activity supports prior evidence that CD8+ cells drive rapid virus escape. These data provide insight into the mechanisms of CD8mediated viral inhibition, and suggest that comparable functional analyses will be important for determining whether similar activities can be induced by T-cell directed vaccine strategies.

AIDS Vaccine 2011

65

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA06.03
The RV144 Vaccine Induced CD4+ T Cells Producing Th1 and Th2 Cytokines with Proliferative Potential

Oral Abstract Session 06: T-Cell Immunity and Immune Escape OA06.04
Association of Timing of HLA B*8101 and B*3910 Mediated p24 Sequence Evolution and Pathways to CTL Escape with Disease Progression
R.S. Ntale1, D.R. Chopera1, N. Ngandu1, d. Debra2, M. Mlotshwa2, C.M. Gray1, K. Mlisana 3, S. Abdool Karim3, C. Williamson1, The CAPRISA 002 Study Team1
1

S. DeRosa1, N. Frahm1, O.D. Defawe1, D. Carter1, E.P. Thomas1, R. Smith1, R. Gottardo1, N. Kochar1, H. Janes1, Y. Fong1, X. Yu1, N.L. Michael2, J.H. Kim2, S. Rerks-Ngarm3, M.J. McElrath1
1 2

Fred Hutchinson Cancer Research Center, Seattle, WA, USA; US Military HIV Research Program, Rockville, MD, USA; 3Thai Ministry of Public Health, Bangkok, Thailand

University of Cape Town, Cape Town, South Africa; 2National Institute for Communicable Diseases, Johannesburg, South Africa; 3Centre for AIDS Programme Research in South Africa, University of KwaZulu-Natal, Durban, South Africa

Background: The RV144 efficacy trial demonstrated moderate reduction in HIV acquisition for participants receiving recombinant canarypox encoding Env, Gag, and Protease, and recombinant gp120 proteins. Methods: PBMC from 10 placebo and 40 vaccine recipients were examined at baseline, Week 26 (two weeks post last vaccination) and Week 52. Intracellular cytokine staining (ICS) measured T cells producing IFN-, IL-2, TNF-, or expressing CD40L, following 6-hour ex vivo stimulation with insert-matched Env and Gag peptide pools. Multiplex bead array measured 12 cytokines in PBMC supernatant following 48-hour stimulation. The CFSE assay measured proliferation. Results: Responses were largely restricted to CD4+ T cells and to Env. CD40L detected the highest response rate by ICS (60% at Week 26, 20% at Week 52), with a median magnitude of 0.1% CD4+ T cells expressing CD40L at Week 26. The Week 26 response rate for IL-2 was 43%, but only 18% for IFN-; both decreased at Week 52 (23% and 13%, respectively). Response rates were similar by multiplex bead array (50% for IL-2 and 14% for IFN- at Week 26), and additional responses of Th2 and regulatory cytokines were detected (30% for IL-4, 44% for IL-5, 53% for IL-13, and 28% for IL-10). Proliferative response rates were lower at Week 26 (16% for Env), but persisted at Week 52 (21%). For all assays, false positive responses were minimal (<5%). Conclusion: In this pilot immunogenicity study, CD4+ T-cell responses were detected in a majority of vaccine recipients, and were polyfunctional including secretion of Th2 cytokines. This, together with expression of CD40L, indicates ability to provide B-cell help. A smaller proportion of vaccine recipients had proliferative T-cell responses, but these appeared more durable, persisting to one year. These assays will be used to examine samples from cases and matched controls and may identify a correlate of protection.

Background: HLAs that are associated with control of vireamia, often select for mutations in Gag p24 in acute/early phases of the HIV1 infection. Methods: To understand the role of viral and host factors in the pathogenesis of subtype C, HIV-1 infection, we timed CTL mediated sequence evolution in TL9 (Gag 180 188) epitope in p24 and compared disease progression in B*8101 (n=5) and B*3910 (n=2) participants from the CAPRISA acute infection cohort. Results: We report that both the B*3910 and B*8101 alleles select mutations in the TL9 epitope, in which either the 182Q or both the 182Q and 186T are replaced by S (TPqDLNtML). However, the timing to sequence evolution differed among these alleles. While both B*3910 participants selected acute/early mutations, four of the five (4/5) B*8101 individuals selected late mutations in the TL9 despite persistent CTL responses targeting this epitope. One B*8101 participant did not develop any restricted sequence evolution in the TL9 epitope by 2.5 years post-infection despite consistent CTL responses. In three of the remaining four individuals, sequence evolution in TL9 was accompanied or preceded with changes in SV9 (Gag 148 156) epitope or amino acids flanking this epitope. The fourth participant developed an E177D mutation in addition to the double mutations in the TL9 epitope. These three mutations were also developed by one of the B*3910 individual but in the acute phase of the infection. Irrespective of HLA, participants who selected both the Q182S and T186S mutations had a slow disease progression profile. Conclusion: These results provide new evidence on the role of the restricting HLA type in the timing of sequence evolution and contribute new information on the constituents of effective CTL responses. In addition, these results further support inclusion of multiple conserved regions of Gag in a vaccine meant to render HIV-1 less fit.

ORAL ABSTRACT SESSIONS

66

AIDS Vaccine 2011

Oral Abstract Session 06: T-Cell Immunity and Immune Escape OA06.05
Evidence of HIV-Specific ADCC Immune Escape
A.W. Chung1, G. Isitman1, M. Navis1, M. Kramski1, R.J. Center1, S.J. Kent1, I. Stratov1
1

Oral Abstract Sessions

OA06.06
Imbalance Between T Helper Type 17 and T Regulatory Cells: Implications for HIV Pathogenesis
D. Li1, J. Chen1, M. Jia1, K. Hong1, Y. Ruan1, H. Liang1, H. Peng1, P. Ma1, Y. Shao1 National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control, Beijing, China
1

University of Melbourne, Parkville, Vic, Australia

Background: Natural killer (NK) cell-mediated antibody dependent cellular cytotoxicity (ADCC) is a potentially important immune mechanism against HIV. No previous work has identified immune escape from ADCC. Rapid evolution of HIV is heavily influenced by focused immune responses directed at particular neutralising antibody or CD8+ T lymphocyte epitopes. Demonstrating viral escape from ADCC responses would be an important contribution to establishing the immune pressure applied by ADCC responses against HIV infection. Methods: Using an ICS ADCC assay for antibody-dependent NK cell expression of IFN and CD107a to overlapping HIV-1 peptides pools, we mapped ADCC epitopes from a cohort of 80 ART nave HIV-positive subjects. We sequenced autologous plasma virus samples across relevant epitopes, identifying variations from consensus subtype B sequence, then compared the ADCC activity induced by consensus and autologous epitopes, including autologous epitope sequences at earlier time points of infection. Glycosylated gp140 AD8 proteins containing ADCC escape mutations were also synthesised and ADCC activity was assayed. Results: 37 linear ADCC epitopes were mapped, primarily in Env; of these, 9 epitopes were shared by 2 or more subjects. Mutational escape from ADCC responses was detected in 8 of the 12 epitopes that we tested. Virus epitope sequences from earlier infection were identical or more similar to consensus. ADCC responses to earlier autologous virus were commonly unable to recognise concurrent virus sequence. ADCC escape was also observed from gp140 ADCC escape mutant proteins compared to wild type gp140 AD8 protein. Conclusion: Loss of ADCC responses induced by epitopes sequenced early in infection strongly indicates that ADCC responses can force mutational escape in HIV-infected subjects. Weaker ADCC activity to gp140 ADCC escape mutant proteins confirm virus escape. This work establishes an additional immune pressure on viral evolution and predicts that ADCC responses induced by vaccination should be capable of reducing virus replication.

Background: T helper 17 (Th17)and CD4+CD25hiFoxp3+ regulatory T (Treg) cells have been described as two distinct subsets from T helper 1 (Th1) and T helper 2 (Th2) cells, their balance may play an important role in the pathogenesis of HIV-1 infection. Methods: 115 untreated chronic HIV-infected individuals and 32 healthy donors were recruited in this study. Peripheral blood mononuclear cells were isolated and stained to characterize the frequencies of Th17 and Treg. 42 individuals including 10 elite controllers were followed up for more than one year, changes of Th17 and Treg frequencies were analyzed over time. Results: Th17 levels with CD4 counts were depleted while Treg were increased in the cohort of 115 HIV-infected individuals, compared to healthy controls (median=0.608% vs 0.938%, 5.150% vs 4.605%; p<0.001, p=0.032), leading to an obvious imbalance of Th17/Treg when compared to the healthy control counterparts (median=0.117 vs 0.194; p<0.001). In the 42 patients, loss of Th17 contrasted with an increased Treg frequencies from Jun 2009 to Oct 2010 were followed longitudinally (median= 0.669% vs 0.603%, 4.765% vs 5.890%; p= 0.078, 0.010). Meanwhile, the elite controller group had comparable Treg, Th17 levels and a Th17/ Treg with healthy controls during the follow-up study, (in 2009, median= 4.555% vs 4.605%, 0.843% vs 0.938%, 0.176 vs 0.194; p= 0.948, 0.635, 0.585; in 2010, median=5.205% vs 4.605%, 0.768% vs 0.938%, 0.140 vs 0.194; p=0.159, 0.220, 0.053). Additionally, we demonstrated that loss of balance between Th17 and Treg could lead to an earlier CD4 T cell decline than Treg or Th17 during the course of HIV infection. Conclusion: The loss of Th17 cells and a reciprocal increase in the Treg cells in HIV-1 chronic infections with no differences observed in elite controllers give prominence to the understanding of HIV-1s pathogenesis and prompt novel considerations for future thoughts on HIV vaccine development.

AIDS Vaccine 2011

67

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA07.01

Oral Abstract Session 07: B Cell Immunology OA07.02


Identification of Amino Acid Residues in HIV-1 Envelope Targeted by Plasma Broadly Neutralizing Antibodies using Evolutionary Models
M. Lacerda1, P.L. Moore2, N. Ngandu3, E.S. Gray2, K. Wibmer2, M. Nonyane2, D. Sheward3, B.T. Korber4, D.C. Montefiori5, C. Williamson3, L. Morris2, C. Seoighe6
1

Bispecific Fusion Antibodies PG9-Ibalizumab (PG9-iMab) and VRC01-Ibalizumab (VRC01-iMab) Exhibit 100% Breadth and Picomolar Potency
C.S. Pace1, R. Song1, D. Franco1, M. Seaman2, D.D. Ho1
1

Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY, USA; 2Beth Israel Deaconess Medical Center, Boston, MA, USA

Background: Ibalizumab, PG9 and VRC01, three of the broadest and most potent monoclonal antibodies against HIV-1 infection described to-date, neutralize 66% - 90% of HIV-1 strains. Here, we created PG9-iMab and VRC01-iMab bispecific antibodies, exploiting ibalizumabs high affinity for domain 2 of CD4 and inherent antiviral activity to create bispecific fusion antibodies that anchor PG9 or VRC01 at the site of viral entry. Methods: Bispecific antibodies were created by fusing PG9 or VRC01 scFv to the N-terminus of the ibalizumab heavy chain. Breadth and potency were assessed to a maximum concentration of 10 g/mL against a well characterized panel of HIV-1 Env pseudotypes (n=118). Results: Addition of PG9 or VRC01 scFv to the N-terminal of ibalizumab was well tolerated; expression and purification yields of bispecific antibodies were comparable to the parent antibodies and had negligible effects on CD4 binding affinity. PG9-iMab and VRC01-iMab neutralized 100% of viruses to at least 50% inhibition (median IC50, 24 pM and 135 pM, respectively) and indeed, PG9-iMab neutralized 97% of viruses and VRC01-iMab neutralized 95% of viruses, to at least 95% inhibition. Most importantly, PG9-iMab and VRC01-iMab each potently neutralized dual PG9- and ibalizumab-resistant viruses and dual VRC01- and ibalizumab-resistant viruses, respectively. PG9-iMab exhibited synergistic potency, neutralizing each virus 100 375 fold and 32 282 fold more potently than PG9 and ibalizumab, respectively (median IQR). In contrast, while potency of VRC01-iMab was enhanced 19 75 fold compared to VRC01, potency was comparable to ibalizumab (3 91 fold). The synergistic activity and ability to neutralize dual-resistant viruses required CD4 anchoring and could not be achieved by simply co-administering PG9 or VRC01 with ibalizumab, which yielded only additive effects. Conclusion: Bispecific ibalizumab antibodies are broad and potent candidates for passive or gene-delivered biologics for prevention and treatment of HIV-1 infection.

University of Cape Town, Cape Town, South Africa; 2AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa; 3Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa; 4Los Alamos National Laboratory/ Santa Fe Institute, Los Alamos/ Sante Fe, NM, USA; 5Department of Surgery, Duke University Medical Center, Durham, NC, USA; 6School of Mathematics, National University of Ireland, Galway, Galway, Ireland

Background: During the course of HIV-1 infection, a small proportion of individuals develop broadly cross-reactive antibodies capable of neutralizing diverse HIV-1 strains. Identification of the epitopes targeted by these antibodies will provide important clues for the design of a preventative vaccine against HIV-1 infection. We have developed an evolutionary model to identify key residues targeted by plasma neutralizing antibodies. Methods: Five HIV-1 subtype C serum samples from the CAPRISA 002 Acute Infection cohort previously shown to have neutralization breadth were tested against a large multiclade panel of 206 pseudoviruses for which gp160 sequences were available. Amino acid residues that influence neutralization sensitivity at ID50 were identified by using Bayes factors (BFs) to compare the fit of a model in which the evolution of the residue correlates with changes in neutralization sensitivity to that of a null model in which the virus evolves independently of the antibody response. Sites with significant BFs were mapped onto the three-dimensional protein structure to identify epitopes on the surface of the envelope glycoprotein. Predictions were tested experimentally by site-directed mutagenesis. Results: We identified between 1 and 4 sites per sample that were strongly associated with neutralization sensitivity (BF>20) and confirmed a subset of the predicted residues experimentally for 4 of the 5 sera. Our model predictions included two sites in the V2 region (166 and 169) that contributed to a trimer-specific PG9/16-like epitope in one subject, a glycan at position 332 in a second individual and the 674 residue in the membrane proximal region of gp41 in a third participant. Conclusion: Our results provide strong support for the use of evolutionary models applied to cross-sectional viral neutralization data to identify novel epitopes recognized by antibodies that confer neutralization breadth. Such epitopes are difficult to discover experimentally or with existing computational tools, thereby demonstrating the utility of our approach.

ORAL ABSTRACT SESSIONS

68

AIDS Vaccine 2011

Oral Abstract Session 07: B Cell Immunology OA07.03


Isolation of a Subset of Novel HIV-1 Broadly Neutralizing Antibodies with Dual Specificity Overlapping the Env Receptor and Coreceptor Binding Sites
Y. Li , S. ODell , X. Wu , S. Schmidt , C. Hogerkorp , Y. Feng , B. Chakrabarti1, R. Wilson1, N. Longo2, N. Doria-Rose3, M. Roederer2, M. Connors3, R. Wyatt1, J. Mascola2
1 2 2 2 2 1

Oral Abstract Sessions

OA07.04
Neutralizing Antibodies Inhibit HIV-1 Transfer from Dendritic Cells to Primary CD4 T Lymphocytes
B. Su1, K. Xu1, M. Peressin2, A. Lederle1, S. Schmidt1, G. Laumond1, C. Moog1, V. Holl3
2

International AIDS Vaccine Initiative/The Scripps Research Institute, La Jolla, CA, USA; 2Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; 3 Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
1

INSERM U748, Universit de Strasbourg, Strasbourg, France; Etablissement Franais du Sang, U725 Cellules Dendritiques, Strasbourg, France; 3Covance CLS SA, Meyrin, Switzerland
1

Background: Both CD4 binding site (CD4 bs) and coreceptor binding site (CoR bs) of HIV-1 gp120 are two of the most conserved functional elements of HIV Env. Elicitation of potent neutralizing antibodies (nAbs) targeting these structural elements with Env-based immunogen has been pursued extensively, with limited success. Our previous study showed that broadly and potent CD4 bs-specific nAbs can be elicited during natural infection at high titers and can be detected in selected patient sera. From the memory B cell repertoire of one such patient, we isolated a subset of CD4 bs-specific nAbs including VRC01, which neutralizes more than 90% of circulating primary virus isolates. From the same patient sera, we detected that there was also a fraction of potent nAbs specific for the Env CoR bs by differential protein adsorption and neutralization analysis. Methods: In this study, we sought to isolate CoR bs-specific nAbs from this patient by sorting with a fluorescence-tagged gp120 possessing a CoR bs knockout mutation (I420R). Results: A series of B cell clones were isolated by the FACS phenotype of WT gp120-high/I420R-low. A subset of these clones was able to neutralize ~ 50% of Clade B tier 2 primary virus isolates, with better potency than that of the typical CoR bs antibodies isolated previously. These clones displayed gp120 binding affinity abrogated by gp120 with both CD4 bs and CoR bs mutations, suggesting its overlapping epitope, which was also supported by cross competition analysis with site-specific ligands. Conclusion: We have isolated a subset of novel CD4/CoR nAbs whose epitopes may directly or functionally overlap with both the HIV gp120 receptor and coreceptor binding sites. Fine epitope mapping and impact on the configuration of the viral Env functional spikes upon antibody binding is under way to define the unique specificity.

Background: Sexual mucosal transmission is the main route of HIV-1 infection worldwide. Immature dendritic cells (DCs), which reside in genital mucosal surfaces, are among the first cells that encounter HIV-1. We have shown previously that in HIV-1 transfer conditions, where HIV-1 loaded immature DCs were cocultured with primary CD4 T lymphocytes, HIV-1 replication was enhanced in DCs. Here, we aim to analyze the kinetic of HIV-1 replication and the ability of HIV-1 specific antibodies to inhibit HIV-1 infection in the co-culture of immature DCs with primary PHA-activated CD4 T lymphocytes. Methods: DCs were generated from purified human blood CD14+ monocytes. After 2 hours of immature DCs incubation with R5 HIV-1 strains, we added autologous primary CD4 T lymphocytes in the presence or absence of HIV-1 specific antibodies. The percentage of infection was quantified by flow cytometry using intracellular p24 viral antigen staining. Results: In this co-culture condition, the early step of HIV replication was more rapid in co-cultured DCs compared to infected DCs without lymphocytes, whereas the kinetic of HIV1 fusion was unchanged. Monoclonal neutralizing antibodies, but not non-neutralizing or inhibitory antibodies, were able to potently inhibit HIV-1 transfer to CD4 T lymphocytes. Moreover, we found a decrease of HIV-1 replication in DCs although neutralizing antibodies or non-neutralizing inhibitory antibodies were added on HIV-1-loaded DCs after two hours. This inhibition of HIV-1 replication in DCs was correlated with DC maturation. Conclusion: These results demonstrate that neutralizing antibodies are able to efficiently inhibit HIV-1 infection of DCs and HIV-1 transfer to primary CD4 T lymphocytes. The efficient inhibition of HIV-1 transfer strengthens the requirement of the induction of HIV-1 specific antibodies directly at mucosal portal of HIV-1 entry. Vaccines that induce such antibody response may prevent the very early dissemination of HIV-1 after its sexual transmission.

AIDS Vaccine 2011

69

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA07.05

Oral Abstract Session 07: B Cell Immunology OA07.06


Influence of NK Cells and NK Cell Receptor Polymorphisms in the Assessment of HIV-1 Neutralization
B.K. Brown1, L. Wieczorek1, G. Kijak2, K. Lombardi1, J. Currier1, M. Wesberry1, C. Ochsenbauer3, J.C. Kappes3, V. Ngauy1, M. Marovich1, N. Michael1, D.C. Montefiori2, V.R. Polonis1
1 2

Induction of Neutralizing Antibody Responses in HIV Infection Is Associated with HIV-Specific CD4 T Cell Responses
M. Mueller1, M. Lindqvist1, D. Soghoian1, S. Ranasinghe1, T. Wrin2, P. Phung2, S. Deeks3, C. Pilcher3, F. Hecht3, H. Streeck1
1

Ragon Institute of MGH, MIT and Harvard, Somerville, MA, USA; 2Monogram Biosciences, San Francisco, CA, USA; 3 University of California, San Francisco, CA, USA
Background: One of the foremost challenges in the development of an HIV-1 vaccine is that HIV-1 targets CD4-T cells, which are critically important in shaping the immune response to infection. It has been demonstrated that T follicular helper (TFH) CD4-T cells are pivotal for the induction of broadly neutralizing antibody (nAb) responses and for the generation of long-lived B-cell memory maturation. By cognate interaction and cytokine secretion, CD4-T cells expressing CD40L can induce class switching and somatic hypermutation in antigen specific B-cells, which is reflected in the expression of activation-induced cytidine deaminase (AID). Nonetheless, the role of TFH cells and their interaction with B-cells in HIV infection is currently unknown. Methods: Ten subjects were identified during acute HIV infection and followed longitudinally over a median of three years. Neutralizing activity of plasma antibodies against a panel of present and past autologous and heterologous viruses was assessed using a single-replication cycle assay in which full-length envelope genes were incorporated into expression vectors (Monogram Biosciences). Phenotypic and functional characteristics of HIV-specific CD4-T cell responses and their impact on AID expression were assessed longitudinally by multiparameter flow cytometry. Results: The breadth of the broadly neutralizing antibody response to heterologous HIV significantly increased over time in each patient. Interestingly, the expansion in heterologous breadth was not only significantly correlated with an increase in AID expression in B-cells, but also with CD4-T cell expression of PD1, a marker that has been associated with TFH cells. Moreover, a positive association between AID expression and gp120-specific CD40L-responses was detected at the earliest time point. Conclusion: Our data strongly indicate that the induction of broadly neutralizing antibody responses is linked to the function and activation of HIV-specific CD4-T cell responses. These findings will be pivotal in efforts to generate neutralizing antibody responses by vaccination strategies.

US Military HIV Research Program, Rockville, MD, USA; Duke University, Durham, NC, USA; 3University of Alabama at Birmingham, Birmingham, AL, USA

Background: Genetic polymorphisms within the killer immunoglobulin receptor (KIR) and Fc gamma receptor genes of natural killer (NK) cells have been shown to influence the containment of HIV replication, both in vivo and in vitro. Peripheral blood mononuclear cells (PBMC) prepared from different donors for in vitro use contain varying numbers of immune cells, including NKs. This study addresses the impact of this variation and of specific genetic polymorphisms on the measurement of neutralization using PBMC from HIVseronegative donors. Methods: Leukopaks (LP) were obtained from 25 donors and PBMC immune cell populations were quantified using multi-color flow cytometry. Viral permissivity and HIV-1 neutralization profiles were assessed using infectious molecular clones (IMC) and intracellular p24 or Renilla luciferase (LucR) expression as endpoints. HIV-1 IMC with LucR (NL-LucR-BaL.ecto and NL-LucR-SF162.ecto) were used in neutralization assays; sCD4 and polyclonal and monoclonal antibodies were tested. CD4+, CD8+ and NK cells were depleted from PBMC using antibody-coated beads. Results: The PBMC NK cell percentages ranged from 0.5-10.0%. When donor PBMC were stratified into quartiles using mean neutralization titer or viral titer rankings, NK cells were found to be elevated in PBMC yielding higher neutralization titers and lower viral growth (p=0.01, Mann-Whitney). KIR3DS1 and a polymorphism for higher affinity (158-Val) in Fc gamma RIIIa, were both associated with PBMC that showed higher neutralization (p=0.009-0.03, depending on the antibody tested). Furthermore, 100% of KIR3DS1+ PBMC fell into the upper 2 neutralization quartiles (p=0.03, Fishers exact test). NK celldepletion decreased neutralization up to 1000-fold, depending upon the PBMC and antibody tested. Conclusion: NK cells, KIR polymorphisms and Fc gamma RIIIa affinity can influence HIV-1 neutralization results. These findings indicate that the LucR-IMC PBMC platform, with antibody continuously present, simultaneously assesses neutralizing and non-neutralizing antibody-dependent HIV-1 inhibition. This may provide the opportunity to delineate the dominant antibody function(s) in polyclonal responses to vaccines.

ORAL ABSTRACT SESSIONS

70

AIDS Vaccine 2011

Oral Abstract Session 07: B Cell Immunology OA07.07


The Effect of HIV-1 Superinfection on the Development of Neutralizing Antibody Breadth Among Female Sex Workers in Mombasa, Kenya
V.C. Cortez1, J. Overbaugh1
1 Fred Hutchinson Cancer Research Center/University of Washington, Seattle, WA, USA

Oral Abstract Sessions

OA07.08 LB
The Thai Phase Iii Clinical Trial (RV144) Induces the Generation of Antibodies That Target a Conserved Region Within the V2 Loop of gp120
N. Karasavvas1, E. Billings2, M. Rao2, J. Currier2, N.L. Michael2, S. Rerks-Ngarm1, P. Pitisuttithum1, J. Kaewkungwal1, V. Ngauy1, S. Nitayaphan1, S. Madnote1, D. Arworn1, J. Kim2, M. de Souza1
1

Background: Identifying naturally-occurring neutralizing antibodies (NAb) that are cross-reactive against all global subtypes of HIV-1 and is an important step toward the development of an HIV vaccine. It was previously shown that NAb breadth is positively associated with viral diversity. Therefore, we hypothesized that superinfected individuals develop a broader NAb response compared to singly-infected individuals as a result of increased antigenic stimulation by two distinct virus strains. Methods: A prospective cohort study was designed with 12 superinfected women each matched to 3 singly-infected women to assess NAb breadth 5 years post-initial infection, when all 12 women had been superinfected for at least 1 year. Breadth was also measured pre-superinfection to control for differences in the antibody responses to the first infection. Breadth scores were calculated for each woman using a panel of 8 viruses representing 4 different HIV-1 subtypes that exhibit a spectrum of neutralization sensitivities, and the resulting data was modeled using conditional Poisson regression. Results: The mean breadth scores of superinfected women were higher compared to singly-infected women at 5 years post-initial infection (5.75 vs. 3.42). Univariate analysis showed that the NAb response of superinfected women was nearly twice as broad as that of singly-infected women at 5 years post-initial infection (RR=1.68, CI: 1.23-2.30, p=0.001). There was also a significant difference in breadth between superinfected and singly-infected women at 1-year prior superinfection (4.30 vs. 3.23). However, adjusting for breadth from 1-year prior superinfection, viral load, and CD4+ T cell count did not substantially change our original estimate (RR=1.51, CI: 1.01-2.25, p=0.04). Conclusion: These data suggest that superinfection elicits a substantial enhancement of the NAb response with regards to coverage and cross-reactivity in the years following reinfection. Individuals who have been superinfected may represent a unique setting to characterize broadly NAbs and the pathways required to elicit them.

AFRIMS, APO, AP, USA; 2U.S. Military HIV Research Program, Rockville, MD, USA

Background: The Thai phase III clinical trial (RV 144) is the only HIV-1 vaccine regimen to show efficacy against acquisition of HIV-1. Antibodies targeting HIV-1 ENV may contribute to preventing infection. We compared the HIV-1 humoral responses induced by the vaccine to those by natural HIV-1 infections. Methods: Antibody responses to gp120 HIV-1 were evaluated using cyclic peptides spanning 42 amino acids (aa) of the V2 and 35 aa of the V3 loop of the ALVAC (vCP1521) Env gp120. Plasma from 40 HIV-1 uninfected trial participants (32 vaccinee/8 placebo recipients) that completed all injections with ALVACHIV/AIDSVAX B/E boost combination and 20 HIV-1 CRF01_AE infected subjects were tested by ELISA. Results: 31/32 (97%) vaccine recipients had antibody responses against V2 at 2 weeks post final immunization (week 26) with a geometric mean end point titer (GMT) of 1100 (range: 2003200).V2 responses were transient, declining to 19% at 28 weeks post last injection (GMT: 110, range: 100-200). The majority of antibody responses targeted the crown of the V2 loop with aa sequence KQKVHALFYKLDIVPI which includes the 47 binding motif LDI that could bind HIV-1 at mucosal surfaces. Unlike the V2 responses, the frequency of V3 responses was modest (18/32, 56%) with a GMT of 185(range: 100-800) at week 26. In comparison, HIV-1 infected individuals had a lower frequency and magnitude of antibody responses to V2: (10/20, 50%) with a GMT of 400 (range: 100-3200) and a higher frequency and magnitude to V3 (19/20, 95%) with a GMT of 3570 (range: 200-12800). Conclusion: The RV 144 vaccine regimen induced antibodies targeting a conserved region of the V2 loop, which includes the 47 binding motif LDI. These observations suggest that V2 antibodies may be a contributing factor in the prevention of HIV1 acquisition.

AIDS Vaccine 2011

71

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA08.01

Oral Abstract Session 08: Animal Models and HIV Transmission OA08.02
CD4+ T Cell Escape in a SIVmac239-Infected Indian Rhesus Macaque Elite Controller with Breakthrough Viremia
J. Giraldo-Vela1, F.A. Norante1, A.T. Bean1, E.J. Leon2, R. Rudersdorf1, N.A. Wilson1, D.I. Watkins1, J.B. Sacha2
1 2

Evidence for CD4 T-Cell Mediated Immune Control of Macrophage Infection in Rhesus Macaques Undergoing CD4+ Lymphocyte Depletion Prior to SIV Infection
A. Ortiz , B. Li , N.R. Klatt , B. Lawson , E. Ryzhova , P. Carnathan1, M. Paiardini1, J. Else1, F. Gonzalez-Scarano4, S.J. Ratcliffe5, J. Brenchley2, J. Estes6, C. Derdeyn1, G. Silvestri1
1 1 2 1 3

University of Wisconsin, Madison, Madison, WI, USA; Oregon Health and Science University, Beaverton, OR, USA

Emory University, Atlanta, GA, USA; 2Laboratory of Molecular Microbiology, National Institutes of Health, Bethesda, MD, USA; 3Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA; 4 School of Medicine, UT Health Science Center at San Antonio, San Antonio, TX, USA; 5Department of Biostatistics, University of Pennsylvania, Philadelphia, PA, USA; 6The AIDS and Cancer Virus Program, The National Cancer Institute, Frederick, MD, USA
1

Background: The role of CD8+ T cells in containment of retroviral infections is well documented by studies of CD8+ T cell-mediated viral escape. It contrast, the immunological role of retrovirusspecific CD4+ T cells remains unclear, particularly in the setting of elite control. Here, we describe viral escape from CD4+ T cells concomitant with viral breakthrough and loss of elite control. Methods: A SIVmac239-infected Indian rhesus macaque maintaining elite control of viremia (<1,000 vRNA copies/mL plasma) spontaneously lost control, experienced breakthrough viremia, and eventually succumbed to AIDS. We performed whole genome sequencing on plasma virus prior to the virus becoming undetectable and immediately following breakthrough and loss of elite control. We then investigated the T cell response directed against areas in the viral proteome exhibiting sequence divergence between the pre- and post-breakthrough time points. Results: Comparison of the pre- and post-breakthrough viral sequences revealed surprisingly few amino acid substitutions post-breakthrough. These substitutions occurred within two previously defined and three previously undescribed CD8+ T cell epitopes, confirming the critical role of CD8+ T cells in control of viral replication. Interestingly, we identified two mutations within Gag, V63A and D205E, which occurred within CD4+ T cell epitopes. Both of these mutations abrogated CD4+ T cell recognition. While the V63A mutation also abrogated recognition of an overlapping CD8+ T cell epitope, the D205E mutation was solely targeted by a CD4+ T cell response. We confirmed that no CD8+ T cell responses were targeting this region. Finally, we demonstrated that viruses bearing the GagD205E mutation escaped CD4+ T cell effector function in vitro. Conclusion: Virus-specific CD4+ T cells play an active role in the containment of retroviruses and are likely essential for the maintenance of low-level viremia. How these CD4+ T cells persist and exert their anti-viral function during retroviral infection remains unclear and warrants further investigation.

Background: The relative contribution of CD4+ T-cells to the control of virus replication during primary SIV infection rhesus macaques (RM) remains incompletely defined. To investigate the relationship between CD4+ T-cells and post-peak decline of viremia, we depleted CD4+ lymphocytes from non-inductive immunological sites of five RMs prior to SIV-infection. Methods: Nine RMs were infected intravenously with SIVmac251. Prior to infection, five RMs were CD4+ lymphocyte-depleted using the Cdf OKT4A HulgG1 Ab. Samples were collected longitudinally from blood, bone marrow, lymph nodes (LNs), and rectal biopsies (RBs). Plasma SIV viremia and cell-associated SIV-DNA were measured by RT-PCR. Cell-associated vRNA in tissues was assessed by dual immunohistochemistry and in situ hybridization. Immune function parameters were measured by multi-color flow cytometry, neutralization assays and ex vivo stimulation assays. Results: At the time of infection, CD4+ lymphocyte-depleted RM showed ten-fold lower CD4+ T cell levels in blood and LNs and normal CD4+ T-cell levels in RBs. After infection, CD4+ lymphocyte-depleted animals exhibited a similar peak of viremia but, in contrast to non-depleted RMs, showed no post-peak virus decline, and progressed more rapidly to AIDS. Importantly, no significant differences were found between depleted and undepleted RMs in terms of (i) availability of target cells; (ii) levels of SIV-specific CD8+ T cell responses; and (iii) SIV-specific humoral responses. Interestingly, peak viral load in depleted animals was associated with a striking increase in virus production by macrophages and dendritic cells. Conclusion: These data indicate an essential role for CD4+ T-cells in mediating post-peak decline of viremia in SIV-infected RMs that may be associated with an ability to suppress virus production by macrophages. The complex balance between CD4+ T-cells as target cells and immune effectors may play a greater than anticipated role in the outcome of vaccine efficacy and should be considered during future vaccine design.

ORAL ABSTRACT SESSIONS

72

AIDS Vaccine 2011

Oral Abstract Session 08: Animal Models and HIV Transmission OA08.03
Decreased Penetration of GFP-Labeled Lentiviral Particles in the Vagina and Endocervix of SIV3Vaccinated Macaques
A. Carias1, M. Anderson1, E. Okocha1, M. McRaven1, W. Li2, R.P. Johnson3, T. Hope1
1

Oral Abstract Sessions

OA08.04
Long-Term Programming of Antigen-Specific Immunity from Gene Expression Signatures in the PBMC of Rhesus Macaques Immunized with an SIV DNA Vaccine
J.D. Boyer1, S.E. Belisle2, J. Yin1, D.J. Shedlock1, A. Dai1, J. Yan1, M.G. Lewis3, H. Andersen3, J.A. Karl4, D.H. OConnor4, A. Khan5, N. Sardesai5, R.E. Palermo6, D.B. Weiner1, M.G. Katze6
1

Northwestern University, Chicago, IL, USA; University of Massachusetts Medical School, Worcester, MA, USA; 3New England Primate Research Center, Harvard Medical School, Southborough, MA, USA
2

Background: Immunization with live attenuated SIV strains, such as SIV3, provide robust protection against vaginal challenge with pathogenic SIV strains. However, the precise contributions of humoral and cellular immune responses, and the anatomic compartment(s) in which virus transmission is interrupted are unknown. To gain insight, we examined that the ability of fluorescent virus particles expressing the SIV envelope to penetrate intact genital epithelium in unvaccinated and SIV3vaccinated macaques. Methods: A total of 11 animals were studied: 8 were vaccinated with SIV3 29 months previously, and 3 were uninfected controls. Three of the SIV3-vaccinated animals were superinfected with SIVmac251 after vaginal challenge one year after vaccination. Subsequently, female macaques were inoculated intravaginally with PA-GFP-gag labeled virions. Genital tracts were removed 4 hours post-inoculation and immediately dissected and snap frozen in OCT media. Tissue specimens were sectioned and stained for cellular proteins, prior to imaging with fluorescent deconvolution microscopy. Results: Within 4 hours, PA-GFP virions were able to penetrate the simple columnar and stratified squamous epithelia in both SIV3-vaccinated and control animals, penetrating up to depths of 50m. When comparing viral penetration depths between the two sample groups, a significant decrease in penetration of viral particles in the vaginal and endocervical mucosa of SIV3vaccinated animals was observed. Conclusion: Our results indicate that in unvaccinated and SIV3vaccinated animals, PA-GFP virions can penetrate to depths where they can interact with target cells in both squamous and columnar epithelium. A decrease in median depth penetration of virus particles in vaccinated macaques suggests vaccination with live attenuated SIV is able to block early events of virus transmission, most likely via the induction of SIV-specific antibody responses. A clearer understanding of the ability of virus-specific antibody responses to interrupt early events of viral transmission at mucosal sites is essential for the development of future HIV vaccine strategies.

University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, University of Washington, Seattle, WA, USA; 3Bioqual, Rockville, MD, USA; 4Wisconsin National Primate Research Center, Madison, WI, USA; 5 Inovio Pharmaceuticals, Blue Bell, PA, USA; 6University of Washington, Seattle, WA, USA
2

Background: While HIV-1-specific cellular immunity is thought to be critical for the suppression of viral replication, the correlates of protection have not yet been determined. Rhesus macaques (RM) are an important animal model for the study and development of vaccines against HIV/AIDS. Our laboratory has helped to develop and study DNA-based vaccines in which recent technological advances including genetic optimization and in vivo electroporation (EP) have helped to dramatically boost their immunogenicity. Methods: In this study, RMs were immunized with a DNA vaccine including individual plasmids encoding SIV gag, env, and pol alone, or in combination with a molecular adjuvant, plasmid DNA expressing the chemokine ligand 5 (RANTES), followed by EP. Along with standard immunological assays, flow-based activation analysis without ex vivo restimulation and highthroughput gene expression analysis was performed. Results: Strong cellular immunity was induced by vaccination which was supported by all assays including PBMC microarray analysis that identified the up-regulation of 563 gene sequences including those involved in interferon signaling. Furthermore, 699 gene sequences were differentially regulated in these groups at peak viremia following SIVmac251 challenge. We observed that the RANTES-adjuvanted animals were significantly better at suppressing viral replication during chronic infection and exhibited a distinct pattern of gene expression which included immune cell-trafficking and cell cycle genes. Furthermore, a greater percentage of vaccine-induced central memory CD8+ T-cells capable of an activated phenotype were detected in these animals as measured by activation analysis. Conclusion: Thus, co-immunization with the RANTES molecular adjuvant followed by EP led to the generation of cellular immunity that was transcriptionally distinct and had a greater protective efficacy than its DNA alone counterpart. Furthermore, activation analysis and high-throughput gene expression data may provide better insight into mechanisms of viral control than may be observed using standard immunological assays.

AIDS Vaccine 2011

73

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA08.05
Compromised NK Cell-Mediated AntibodyDependent Cellular Cytotoxicity in Chronic SIV Infection

Oral Abstract Session 08: Animal Models and HIV Transmission OA08.06
Cytotoxicity Capacity of SIV-Specific CD8+ T Cells Against Primary Autologous Targets Correlates with Immune Control in SIV-Infected Rhesus Macaques
D. Mendoza1, S. Johnson1, B. Peterson1, N. Doria-Rose1, G. Franchini2, D. Schneider3, M.T. Trivett3, C.M. Trubey3, V. Coalter3, D.I. Watkins4, J.D. Lifson3, S.A. Migueles1, M. Connors1
1 National Institute of Allergy and Infectious Diseases/ National Institutes of Health, Chevy Chase, MD, USA; 2 Vaccine Branch, National Cancer Institute/National Institutes of Health, Bethesda, MD, USA; 3AIDS and Cancer Virus Program, SAIC-Frederick, Inc., Frederick, MD, USA; 4Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA

X. He1, Z. Luo1, M. Jia1, Y. Li2, Z. Cong3, D. Li1, H. Liang1, Y. Shao1


1

National Center for AIDS/STD Control and Prevention in China, Chinese Center for Disease Control, Beijing, China; 2 College of Life Science, Nankai University, China; 3Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, China
Background: Increasing evidence indicates that antibodydependent cellular cytotoxicity (ADCC) contributes to control of HIV/SIV infection. However, virtually nothing is known about the ADCC function of Nature killer cells in non-human primate models. Efforts to get a clearer understanding of the specific role NK cells play in rhesus macaques will be helpful in evaluation of vaccines in non-human primate models. Methods: The antibody-coated target cells were used to measure the ADCC function of macaque NK cells. The ADCC responses were analyzed by NK cell cytokine secretion and degranulation through ICS-based assay. All data were collected on BD FACS Aria. Results: To identify the differences of NK cell-mediated ADCC function between naive and chronic SIV-infected macaques, some cytokines were measured in respond to Fc target cells. It was obvious to notice the downregulation of CD107a expression (P= 0.0203) as well as decreased secretion of IFN- (P= 0.0180) and TNF- (P= 0.032) in NK cells from SIV-infected group compared with naives. The greatly reduced CD16+ NK subset in macaques with SIV infection (P=0.0209) was the reason of impaired NK cell function, cause there was a direct association between CD16 expression and cytokine production in NK cells, including CD107a(P=0.0204), IFN-(P=0.0231), TNF-(P=0.0413). The loss of CD16 MFI following stimulation, which was used to quantify the Fc-mediated activation of NK cell, was detected much lower in SIV-infected group (P= 0.0036). Conclusion: In chronic SIV-infected group, we could see the significant compromised ADCC function and its close relationship with CD16 expression on NK cells: the increased levels of CD16 allow NK cells to better respond to Fc target cells and this marker could be served as sensor for ADCC function of NK cells in macaques. These data offer new insights into the role of NK cells as an important bridge between innate and adaptive immunity in non-human primate models.

Background: HIV-specific CD8+ T cells of long-term nonprogressors / elite controllers (LTNP/EC) exhibit extraordinary per-cell cytotoxic capacity against HIV-infected CD4+ T cells, in contrast to those of progressors. In this study, we explore whether our previous findings can be extended to the SIV-rhesus macaque (RM) model by measuring SIV-specific cytotoxic capacity of CD8+ T cells against SIV-infected targets. Methods: Cytotoxicity and IFN- production of SIV-specific CD8+ T cells were examined following 6-day incubation with autologous SIVmac251-infected telomerase-transduced CD4+ T cell clone targets. SIV-specific CD8+ T cell cytotoxic responses were measured at 1 hour by flow cytometric detection of active granzyme (Gr) B delivery to live targets and infected CD4 elimination (ICE). Results: Fifteen SIV-infected RM with different extents of control of viral replication have been evaluated. Data collection is ongoing with laboratory personnel blinded to RM disease status. LTNP/EC RM had higher median delivery of active GrB to targets (43.5% vs. 18.5%, p=0.04), and higher median ICE than progressors (67.3% vs. 24.4%, p=0.009). There was a significant correlation between ICE and viral load (R= -0.55, p=0.03), and between GrB delivery and ICE (R=0.86, p<0.0001). Conclusion: Lytic granule loading of CD8+ T cells and efficient delivery of active GrB to SIV-infected targets are associated with control of SIV infection in rhesus macaques, consistent with observations of HIV infection in humans. These findings suggest ICE is a correlate of control of viral replication in chronic SIV infection. They also suggest the use of cytotoxic capacity as a predictor of immunologic control in the vaccine setting should be tested.

ORAL ABSTRACT SESSIONS

74

AIDS Vaccine 2011

Oral Abstract Session 08: Animal Models and HIV Transmission OA08.07
Live Attenuated Influenza-SIV Vaccines: Efficacy Against Mucosal Challenge in Macaques
S.J. Kent1, S. Jegaskanda1, J. Reece1, T. Ameresena1, A. Sexton1, C. Fernandez1, J. Stambas1, A. Brooks1, R. De Rose1
1

Oral Abstract Sessions

OA08.08 LB
Reduction of Founder Virus in Vaccinated Monkeys After Mucosal SIV Challenge
J.B. Whitney1, M. Rolland2, M. Lacerda3, P.T. Hraber4, A. DeCamp5, C. Luedemann1, S.S. Rao6, J.R. Mascola7, B. Korber4, G. J. Nabel7, P. Gilbert5, C. Seoighe8, N.L. Letvin1
1

University of Melbourne, Parkville, Australia

Background: Attenuated Influenza-HIV vaccines are attractive since they are easy to deliver, induce mucosal immunity and have established manufacturing processes. Methods: We assessed Influenza-SIV constructs expressing 3 SIV CTL epitopes restricted by Mane-A*10 engineered into the neuraminidase stalk. Vaccines were administered primarily intranasally to 15 Mane-A*10+ pigtail macaques. Results: Prime-boost vaccinations with H1N1 and H3N2 constructs were well tolerated via the respiratory route and all animals seroconverted to Flu, had Flu RNA recovered early after vaccination, and generated Flu-specific CTL responses. Vaginal inoculations did not result in Flu seroconversion despite nonoxynl-9 pretreatment. SIV-specific responses were low after vaccination but expressed the mucosal homing marker a4b7 and rose dramatically after intravaginal SIV exposure. SIV RNA levels were however, high after challenge and rapid serial escape at the SIV CTL epitopes was observed. Indeed, immune escape was faster in vaccinees than controls (p = 0.02) and the SIV specific CTLs rapidly became dysfunction after challenge in comparison to Flu-specific CTLs. Conclusion: Although live attenuated influenza vaccines prime SIV-specific CTL immunity, broader immunity will be required to maintain viral control. We are currently engineering recombinants expressing a larger suite of HIV/SIV antigens in vectors that replicate more effectively in primates to induce more broadly based T and B cell immunity.

Beth Israel Deaconess Medical Center and Harvard University, Boston, MA, USA; 2U.S. Military HIV Research Program (MHRP), Rockville, MD, USA; 3School of Mathematics, Statistics and Applied Mathematics,NUI, Galway, Ireland; 4Los Alamos National Laboratory, Los Alamos, NM, USA; 5The Statistical Center for HIV/AIDS Research and Prevention, Seattle, WA, USA; 6Vaccine Research Center, NIAID, Bethesda, MD, USA; 7Vaccine Research Center (NIAID), Bethesda, MD, USA; 8School of Mathematics, Statistics and Applied Mathematics, Galway, Ireland
Background: Background: A key criteria in the NHP evaluation of candidate vaccines is determining the presence of any vaccineinduced effects on the genotype or number of founder strains acquired during repeated low-dose mucosal challenges. We have undertaken studies in a cohort of 167 monkeys to determine the presence of any vaccine-induced sieving effects, or differences in minimum number of SIV founders initiating infection in vaccinated or sham-control animals. Methods: Methods: 167 rhesus monkeys received either an experimental DNA prime Ad5 boost regimen, or sham vaccination. All monkeys were challenged intra-rectally with a repeat lowdose SIVE660 or SIVmac251 challenge 40 weeks after the last vaccination. We amplified the envelopes of blood-associated SIVE660 at the earliest point after viral infection using single genome amplification. Full-length SIV envelope sequences were subject to extensive bio-informatic and statistical analysis to evaluate the minimum number of virus acquired during the infection of each monkey, and if vaccine-induced sieving effects were present. Results: Results: Overall, evidence of vaccine-induced sieving was observed in virus sequences derived prior to the peak of virus replication. However, this sieving was most pronounce in monkeys expressing the Mamu A*01 allele. We also observed a significant reduction in the minimum number of founder virus initiating infection in vaccinated monkeys with permissible host genotypes. We also observed altered levels of mutation in virus sequences isolated from vaccine recipients. Conclusion: Conclusions: Evidence of vaccine-induced effects can be observed at the earliest times after virus challenge and can impact viral diversity post acquisition. Importantly, vaccination impacts the number of acquired viral lineages, but significant levels of specific immunological sieving was observed only in a subgroup of vaccinees. These data highlight non-specific immune mechanisms of vaccine-interrupted virus acquisition. Moreover, the inclusion of a quantitative endpoint for founder/ transmitted virus should be considered for future evaluations of candidate vaccine efficacy.

AIDS Vaccine 2011

75

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA09.01
T-Cell Responses Induced by Immunization with Ad35-HIV-1 Vaccines Are Broad, Durable, Polyfunctional and Can Mediate Inhibition of HIV
J.H. Cox , P. Hayes , E. Cormier , D. Gill , J. Kopycinski , A. Spentzou2, L. Hachaambwa3, C. Bunce3, E. Sayeed1, A. Lombardo1, W. Komaroff1, K. Loughran5, B. Barin5, L. Dally5, J. Ackland1, T. Tarragona4, M. Keefer3, J. Excler1, P. Fast1, J. Gilmour2
1 2 2 2 2 1

Oral Abstract Session 09: Pre-Clinical and Early Clinical Trials OA09.02
ADCC Titers to HIV-Infected Cells Are Detectable in the Majority of Vaccine Recipients in the RV144 Trial
M.D. Alpert7, J.D. Harvey7, W.J. Neidermyer7, Y. Huang1, D. Morris1, L. Harris1, H. Gao2, D.C. Montefiori2, P. Pitisuttithum3, J. Kaewkungwal3, S. Nitayaphan4, S. Rerks-Ngarm5, N.L. Michael6, J.H. Kim6, D.T. Evans7
1 SCHARP, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 2Duke University Medical Center, Durham, NC, USA; 3Mahidol University, Bangkok, Thailand; 4Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 5 Ministry of Public Health, Nonthaburi, Thailand; 6Walter Reed Army Institute of Research, U.S. Military HIV Research Program, Rockville, MD, USA; 7Harvard Medical School, Southborough, MA, USA

International AIDS Vaccine Initiative (IAVI), New York, NY, USA; 2IAVI Human Immunology Laboratory, London, United Kingdom (Great Britain); 3University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; 4International AIDS Vaccine Initiative, Human Immunology Laboratory, London, United Kingdom (Great Britain);5The EMMES Corporation, Rockville, MD, USA

Background: Broad, robust, polyfunctional T cells able to mediate functional inhibition and/or killing of HIV-infected CD4 targets are critical for immunological control of HIV replication. Ad35 replication-defective vectors may be able to induce these responses and circumvent pre-existing immunity to common human adenoviruses. Methods: A Phase I trial of Ad35 expressing HIV-1 subtype A gag, RT, integrase, nef (GRIN as a fusion protein) and envelope (ENV) genes or GRIN alone was conducted in 56 healthy HIV-uninfected, Ad35 antibody negative adults in the U.S. The vaccines were administered intramuscularly at 0 and 6 months at 2x109, 2x1010 or 2x 1011 viral particles (vp) for groups A-C, respectively. Group D received Ad35-GRIN alone at 1x1010 vp. Immunogenicity was assessed by IFN-g ELISPOT, polychromatic flow cytometry, viral inhibition, ENV ELISA and Ad35 neutralization. Results: IFN-g ELISPOT responses to any vaccine antigen in groups A-D were detected in 50, 56, 70 and 90% and in 75, 100, 88 and 86% of vaccine recipients post 1st (Vac.1) and 2nd (Vac.2) vaccination. The geometric mean spot forming cells/ million PBMC to any antigen was 101-122 across groups A-C and 145185 in group D after Vac.1 and Vac.2, with an increase of 0.34 log after Vac.2. Breadth increased from a median of 2 (Vac.1) to 4 (Vac.2) proteins out of 5. CD4 and CD8 T-cell responses were polyfunctional. CD8 T-cell-mediated inhibition of HIV was detected in 61% (Vac.1) and 52% (Vac.2) of vaccinees across multiple HIV-subtypes and dose dependent. Env antibodies were detected in all vaccinees in groups A-C. Ad35 neutralizing titers were low even after Vac.2. Conclusion: HIV-specific cellular responses were seen in the majority of volunteers immunized with Ad35-GRIN/ENV or Ad35-GRIN, the response rate, breadth and magnitude of the response increased after the second dose. T-cell responses were polyfunctional and inhibition of HIV was seen.

Background: A modest reduction in the rate of HIV-1 infection was observed among vaccine recipients in the RV144 trial who received a prime-boost vaccine regimen with recombinant canarypox and soluble gp120. Whereas virus-specific CD8+ T cell responses were largely undetectable, antibodies capable of binding to gp120 were detectable in nearly all vaccinated subjects. However, these antibodies were unable to neutralize primary HIV-1 isolates, suggesting that antibodies may have contributed to protection by non-neutralizing effector mechanisms. Methods: To determine if vaccinated subjects made antibodies capable of directing antibody-dependent cell-mediated cytotoxicity (ADCC), we used a novel assay to measure ADCC titers against HIV-infected cells in pilot samples from the RV144 trial. This assay is based on the killing of an HIV-infected CD4+ T cell line by a CD16+ NK cell line in the presence of serial dilutions of plasma, thus allowing ADCC titers to be measured against virus infected cells expressing the native, oligomeric conformation of the HIV-1 envelope glycoprotein. Using this approach, ADCC titers in blinded plasma samples from 80 vaccine recipients and 20 placebo controls were measured at baseline (week 0) and at two weeks after the last boost (week 26) against target cells infected with the HIV-1 subtype AE isolate 92TH023. Results: Comparison of the response rate for the vaccine group versus the placebo group revealed that 76.2% (61/80; 95% CI 66%, 84%) of the vaccinated subjects versus 10.0% (2/20; 95% CI 3%, 30%) of the placebo controls had detectable ADCC titers against HIV-1 92TH023 infected cells at week 26 (Fishers exact test, P=6.7e-8). ADCC titers were also significantly higher at week 26 than at week 0 among the vaccine recipients (McNemars exact p-value, P=3.9e-12). Conclusion: These results demonstrate that the majority of vaccinated subjects in the RV144 trial made antibodies capable of directing ADCC against HIV-infected cells.

ORAL ABSTRACT SESSIONS

76

AIDS Vaccine 2011

Oral Abstract Session 09: Pre-Clinical and Early Clinical Trials OA09.03
V2-Reactive Antibodies in RV144 Vaccinees Plasma
S. Zolla-Pazner1, T. Cardozo1, A. deCamp2, B. Haynes3, J. Kim4, X. Kong1, N. Michael4, S. Rerks-Ngarm5, C. Williams1
1

Oral Abstract Sessions

OA09.04
Surface Plasmon Resonance Analysis of Antigp120 V2-Specific IgG Antibodies Generated in the RV144 Thai Trial
E.A. Billings1, N. Karasavvas2, M.S. de Souza2, J. Currier1, P. Pitisuttithum3, J. Kaewkunwal3, S. Nitayaphan4, P.B. Gilbert5, G.D. Tomaras6, S.B. Zolla-Pazner7, B.F. Haynes6, N.L. Michael8, S. Rerks-Ngarm9, J.H. Kim8, M. Rao8
1

NYU School of Medicine, New York, NY, USA; 2Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 3Duke University School of Medicine, Durham, NC, USA; 4US Military HIV Research Program/Henry M. Jackson Foundation, Rockville, MD, USA; 5Ministry of Public Health, Nonthaburi, Thailand

Background: Initial studies suggested that V2 antibodies (Abs) played a protective role in RV144. Pilot experiments were needed to quantify and characterize anti-V2 Abs in plasma of RV144 participants. Methods: One hundred coded RV144 pilot study specimens were analyzed by ELISA using plasma obtained before immunization (visit 1), and 2 and 28 weeks after the last immunizing dose (visits 8 and 9, respectively). Results were decoded after submission and analysis of data. Results: [a] At visit 1, 0/20 placebo and 3/80 vaccinee specimens diluted 1:20 reacted with gp120JR-FL. At visit 8, 0/20 and 80/80 were reactive. Plasma were not reactive vs. bovine serum albumin. [b] At visit 1, 0/20 placebo and 1/80 vaccinee specimens diluted 1:20 reacted with a V1V2HxB2-gp70 fusion protein. At visit 8, 1/20 and 76/80 (95%) were reactive. [c] Reactivity was tested against four 21-mer linear V2 peptides spanning the 47 binding motif, chosen to represent: the most polar sequence found in V2 loops with 38 amino acids (polar-38); the most common sequence of V2s with 40 AAs (common-40); the most polar sequence in V2 loops with 40 AAs (polar-40); and the consensus sequence of V2 loops with 40 AAs (consensus-40). Activity in plasma diluted 1:100 showed differential reactivity with the peptides: polar-38 polar-40 > consensus > common-40. For visit 8, 1/20 placebo and 47/80 (59%) vaccine specimens were reactive with polar-38. Shorter linear peptides were poorly reactive. Notably, by visit 9, the reactivity to gp120JR-FL, V1V2HxB2-gp70, and all V2 peptides was markedly reduced in all vaccinees plasmas. Conclusion: Anti-V2 Abs were elicited by the RV144 immunization protocol. Reactivity was highest 2 weeks after the last boost, and was significantly diminished 26 weeks later. V2-specific reactivity was demonstrable with a fusion protein containing the entire V1V2 loop and with 21-mer linear V2 peptides spanning the 47 binding motif.

U.S. Military HIV Research Program, Henry Jackson Foundation, Rockville, MD, USA; 2AFRIMS, Bangkok, Thailand; 3 Mahidol University, Bangkok, Thailand; 4Royal Thai Army, Armed Forces Research Institute of Medical Sciences, Thailand; 5Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 6Duke University School of Medicine, Durham, NC, USA; 7NYU, New York, NY, USA; 8U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Rockville, MD, USA; 9Ministry of Public Health, Bangkok, Thailand
Background: The RV144 HIV-1 prime-boost vaccine trial showed a modest level of protection with a vaccine efficacy of 31.2%. The reduced rate of viral acquisition could be due to the induction of envelope-specific antibodies. The V2 loop of HIV-1 gp120 has been implicated as a binding partner of integrin 47. Therefore, a subset (pilot study) of 40 plasma samples was analyzed for IgG antibodies specific to cyclic V2 and cyclic V3 peptides derived from the gp120 subunit of HIV-1 92TH023 envelope protein. Methods: Biacore analyses of 40 plasma samples (32 vaccinee, 8 placebo recipients) from the RV144 vaccine trial before vaccination (visit 1) and 2 and 28 weeks after the last immunization (visits 8, and 9, respectively) were conducted. Cyclic peptides (V2, V2 variants, and V3) were coupled to Biacore sensor chips. Diluted plasma samples (1:50) were injected over the chip surface followed by anti-human IgG antibody. Results: V2-specific IgG antibodies were present in 0/32 from visit 1 and 31/32 (96.8%) vaccinees from visit 8. Vaccinee plasma showed binding to the 42 amino acid cyclic peptide containing the 47 binding motif. Binding was greatly diminished when that region was scrambled. There was significant reduction in V2-specific IgG antibodies in all of the vaccinee plasma samples by visit 9. The RV144 samples analyzed had extremely low or no antibodies reactive with cyclic V3 peptide. In contrast to vaccinees, plasma samples (1:50 dilution) from individuals infected with clade A/E from the same geographical region had >3-fold more anti-V3 antibodies compared to anti-V2 antibodies. Conclusion: Our results support the conclusion that the V2 loop of gp120 as presented in the context of the RV144 vaccine is more amenable to induction of V2-specific antibodies when compared to a natural infection. The majority of the V2-specific antibodies were directed against the region containing the 47 binding motif.

AIDS Vaccine 2011

77

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA09.05
Polyfunctional CD4/CD8 HIV-Specific Cytokine Responses and Presence of Cytolytic Markers in Swedish Vaccinees Immunized with HIV-1 DNA and HIV-1 MVA
K. Godoy-Ramirez1, C. Nilsson2, K. Karln1, T. Tecleab1, M. Marovich3, B. Hejdeman4, B. Wahren2, E. Sandstrm4, G. Biberfeld2
1

Oral Abstract Session 09: Pre-Clinical and Early Clinical Trials OA09.06
PedVacc001: Safety and Immunogenicity of a Candidate HIV-1 Vaccine,MVA.HIVA, Administered to Healthy Gambian Infants Born to HIV-1/2Uninfected Mothers
M.O. Afolabi1, J. Ndure1, J. Mueller1, V. Thomas1, M. Shams-Rony1, N. Borthwick2, A. Bridgeman2, A. Black2, M. Reilly3, W. Jaoko4, G. John-Stewart5, G. Ambler5, B. Lohman-Payne5, T. Hanke2, K.L. Flanagan1
1 Medical Research Council Laboratories, The Gambia, Banjul, Gambia; 2The Jenner Institute, University of Oxford, Oxford, United Kingdom (Great Britain); 3Karolinska Institutet, Stockholm, Sweden; 4Kenya AIDS Vaccine Initiative, University of Nairobi, Nairobi, Kenya; 5Departments of Medicine and Global Health, University of Washington, Seattle, WA, USA

Swedish Institute for Communicable Disease Control, Stockholm, Sweden; 2Swedish Institute for Communicable Disease Control and Karolinska Inst, Stockholm, Sweden; 3US Military HIV Research Program, Walter Reed Army Institute of Resear, Rockville, MD, USA; 4Venhlsan, Karolinska Institutet, Sdersjukhuset, Stockholm, Sweden
Background: We have monitored polyfunctional HIV-1-vaccineinduced responses as measured by cytokine and cytolytic marker expression. Healthy volunteers were immunized at 0, 1 and 3 months with DNA plasmid expressing gp160 of HIV-1 subtypes A, B and C; revB; p17/p24 gag A and B and RTmut B. At nine months they were boosted with a heterologous MVA expressing HIV-1 env, gag, pol of CRF01_AE. Approximately 3 years after the first HIV-MVA vaccination, 24 volunteers were given a second HIV-MVA boost. Methods: HIV-Gag specific immune responses were assessed by IFN-gamma-ELISpot and by 8-colour ICS for expression of cytokines (CD3/CD4/CD8/IFN-gamma/IL-2/TNF-/MIP1-/VIVID) using fresh cells, and for expression of cytolytic markers (CD3/ CD8/IFN-gamma/MIP1-/CD107/Perforin/Granzyme B/VIVID) using cryopreserved cells. Results: Two weeks after the second HIV-MVA vaccination, 19 (83%) of 23 evaluable vaccinees were reactive in IFN-gammaELISpot; 18 to Gag and 11 to Env peptides. The ICS revealed CD4 and/or CD8 Gag-specific reactivity in 15/23 (65%) vaccinees. Polyfunctional CD4+ and CD8+ T cell Gag-specific responses were detected in 7/23 (30%) vaccinees, all of whom had IFNgamma-ELISpot Gag reactivity >175 SFC/million PBMC. Within the CD8+ T-cell compartment approximately 25% of the responding cells were dual functional and 60% polyfunctional, predominantly expressing IFN-gamma/MIP1-, IFN-gamma/ TNF-/MIP1- and IFN-gamma/IL-2/TNF-/MIP-1. Among the responding CD4 T-cells approximately 25% were dual functional and 60% polyfunctional, predominantly expressing IL-2/TNF-, IFN-gamma/IL-2/MIP1- and IFN-gamma/IL-2/TNF-/MIP1-. Conclusion: A second HIV-MVA boost given approximately 3 years after the initial HIV-DNA/ HIV-MVA vaccinations gave rise to polyfunctional immune responses as measured by the production of cytokines and cytolytic markers. The data supports further exploration of this vaccine concept.

Background: The development of a safe and effective HIV-1 vaccine remains a global priority especially in prevention of mother to child transmission of HIV-1 during breastfeeding. Our vaccine platform consists of recombinant BCG prime-recombinant modified vaccinia virus Ankara (MVA) boost. This study is the first stage and evaluates the safety and immunogenicity of MVA. HIVA, a candidate HIV-1 vaccine, among healthy Gambian infants born to HIV 1/2 uninfected mothers. Methods: Sixty-five mothers were sensitized on delivery of healthy babies at Sukuta Health Centre, The Gambia. Sixty-two of whom consented to HIV voluntary counseling and testing and all the mothers (100%) had negative HIV test. Forty-eight of the eligible infants were randomized into vaccine and no-treatment control group at 20 weeks of age. The vaccine group received one dose of 5 x 10^7 pfu of MVA.HIVA administered intramuscularly. The safety of MVA.HIVA was determined by comparing adverse events, reactogenicity, biochemical and haematological data of vaccine and control groups. Immunogenicity was measured by interferon (IFN)- ELISPOT assay on peripheral blood mononuclear cells. Results: The mean Haemoglobin (Hb), White Blood Cell count (WBC), Alanine transaminase (ALT) and Creatinine at prevaccination and postvaccination visits were within acceptable normal ranges. Adverse events observed included fever, excessive crying, cough, vomiting, bilateral eye discharge, diarrhea and poor weight gain. All study infants had negative HIV antibody tests at 28 weeks and no severe adverse events or suspected unexpected serious adverse reactions were reported. Preliminary cultured IFN- ELISPOT analysis showed induction of HIV-1specific T-cell responses in about 40% of the vaccinees. Conclusion: We have shown that MVA.HIVA is safe and immunogenic in healthy Gambian infants. A parallel study PedVacc002 of MVA.HIVA administered to infants born to HIV1-positive mothers is ongoing in Kenya.

ORAL ABSTRACT SESSIONS

78

AIDS Vaccine 2011

Oral Abstract 10: B Cell Vaccine Concepts OA10.01


Eliciting Broadly Neutralizing Antibodies Against HIV-1 Using a Novel Antigen Containing gp41 Membrane-Proximal External Region
D. Han3, H. Habte3, Y. Qin3, K. Takamoto1, C. Labranche2, D. Montefiori2, M. Cho3
1 2

Oral Abstract Sessions


OA10.02
Neutralizing Antibody Responses Induced with V3-Scaffold Protein Boosts Following Priming with gp120 DNA
S. Zolla-Pazner1, X. Kong1, X. Jiang1, T. Cardozo1, A. Nadas1, S. Cohen1, M. Totrov2, M.S. Seaman3, S. Wang4, S. Lu4 NYU School of Medicine, New York, NY, USA; 2Molsoft, LLC, La Jolla, CA, USA; 3Beth Israel Deaconess Medical Center, Boston, MA, USA; 4University of Massachusetts Medical School, Worcester, MA, USA
1

Albert Einstein College of Medicine, New York, NY, USA; Duke University, Durham, NC, USA; 3Iowa State University, Ames, IA, USA

Background: A major roadblock in developing an AIDS vaccine is the difficulty in eliciting broadly neutralizing antibodies (BRNabs). The membrane-proximal external region (MPER) of gp41 is highly conserved and targeted by BR-Nabs 2F5 and 4E10. Thus, it is an attractive target for vaccine development. However, many past attempts to elicit BR-Nabs using MPER-based antigens have failed. Here, we report a unique, comprehensive vaccine strategy that uses a structurally rigid mini-protein based on the MPER and an adjuvant/antigen-delivery platform that can elicit BR-Nabs in rabbits. Methods: We generated a trimeric mini-protein that is structurally rigid, yet efficiently recognized by 2F5, 4E10 and Z13e1. It is based on M group consensus sequence and contains the C-terminal 54 a.a. of gp41 ectodomain (gp41-54Q), which includes the HR2 and the MPER. A 6xHis tag at the C-terminus was used to attach gp41-54Q to Zn-chitosan, which served as an adjuvant and antigen depot. Rabbits were immunized subcutaneously 3-4 times. Antibody responses were evaluated by ELISA and pseudovirus neutralization assays. Results: The mini-protein was highly immunogenic, eliciting antibody titers greater than 1x107. More importantly, antisera exhibited potent, broadly neutralizing activity against Tier 1 clade B and C viruses. Antisera also exhibited weak, but detectable, neutralizing activities against a multi-subtype panel of Tier 2 viruses. Results from a multitude of analyses suggested that we are eliciting a novel neutralizing antibody that is different from 2F5 or 4E10. Conclusion: We have successfully generated a soluble, structurally rigid mini-protein containing gp41 MPER that is able to induce potent, BR-Nabs against HIV-1 in rabbits. These results represent a significant breakthrough in the HIV-1 vaccine field. Considering that the neutralizing antibody we are eliciting is different from 2F5 and 4E10, our study opens up a new avenue of AIDS vaccine research.

Background: The V3 loop is one of the known targets for neutralizing antibody (NAb) responses, and V3-scaffold fusion proteins used as a boost after gp120 DNA priming were previously shown to induce NAbs in rabbits. In the current study, we evaluated whether the breadth and potency of NAb responses could be further improved when boosted with rationally designed and/or double combinations of V3-scaffold immunogens after gp120 DNA priming. Methods: Rabbits were primed with codon-optimized clade C gp120 DNA and then boosted with one of five V3-cholera toxin B fusion proteins (V3-CTBs) or with various double combinations of these V3-CTBs. The V3 inserts in these V3-CTB protein immunogens were rationally designed to present epitopes shared by the majority of global isolates. Immune sera were assessed for (a) neutralization of Tier 1 and 2 pseudoviruses (psVs) infecting TZM.bl cells, and (b) neutralization of PBMC-grown primary isolates infecting TZM-bl cells, the latter being a far less sensitive assay than the former. Results: The V3C-CTB immunogen gave the most broad and potent neutralizing Ab response of the five V3-CTB immunogens tested individually. Generally, the double combinations of V3CTB boosts gave better breadth and/or potency than did boosts with single V3-CTBs. With the double combination boosts, neutralization was achieved against 9/9 Tier 1 psVs from clades AG, B and C, and against 7/14 psVs from the clade B and C standard psV panels. Similarly, neutralization was achieved with 7/15 primary isolates from clades A, AG, and B. Conclusion: The data demonstrate that priming with gp120 DNA followed by boosts with V3-scaffold immunogens effectively elicits cross-clade NAbs.

AIDS Vaccine 2011

79

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA10.03
Phase I Proof of Principle: Immunization with Virosome-gp41-Derived Antigen Induces Mucosal Antibodies with Antiviral Properties
G. Leroux-Roels1, C. Maes2, F. Clement2, F. van Engelenburg3, M. van den Dobbelsteen3, R. Zurbriggen4, M. Amacker5, M. Adler5, M. Spengler5, J. Detmers5, F. Anjuere5, L. Lopalco7, D. Tudor8, A. Drillet8, M. Bomsel8, A. Chalifour9, S. Fleury9

Oral Abstract 10: B Cell Vaccine Concepts OA10.04


DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660
R. Jalah1, V. Patel1, V. Kulkarni1, A. Valentin1, C. Alicea1, M. Rosati1, A. von Gegerfelt1, N.Y. Sardesai2, J. Bess, Jr.3, J.D. Lifson3, B. Keele3, R.R. Amara4, H.L. Robinson5, V.M. Hirsch6, Y. Guan7, D. Venzon8, D.C. Montefiori9, B.K. Felber1, G.N. Pavlakis1 National Cancer Institute at Frederick, Frederick, MD, USA; Inovio Pharmaceuticals, Inc., Blue Bell, PA, USA; 3SAICFrederick, Inc., National Cancer Institute at Frederick, Frederick, MD, USA; 4Yerkes National Primate Research Center, Atlanta, GA, USA; 5GeoVax, Inc., Smyrna, GA, USA; 6 National Institutes of Allergy and Infectious Diseases, Bethesda, MD, USA; 7Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA; 8 National Cancer Institute, Bethesda, MD, USA; 9Duke University Medical Center, Durham, NC, USA
1 2

Faculty of Medicine, Ghent University, Ghent, Belgium; CEVAC, Ghent University and Hospital, Ghent, Belgium; 3 Kinesis Pharma B.V., Breda, Netherlands; 4Pevion Biotech Ltd, Ittigen, Switzerland; 5Chimera Biotec, Dortmund, Germany; 6INSERM UMR U634, Facult de Mdecine Pasteur, Nice, France; 7Fondazione Centro San Raffaele del Monte Tabor Immunbiology of HIV, Milan, Italy; 8Institut Cochin, CNRS UMR8104/ INSERM U567, Paris, France; 9Mymetics Corporation, Epalinges, Switzerland
1 2

Background: The ideal prophylactic vaccine against HIV-1 sexually transmitted should prevent the entry and early infection of HIV-1 at mucosal sites. We have previously demonstrated that mucosal IgAs/IgGs induced by vaccination with influenza virosomes harboring HXB2 rgp41 and modified-P1, a lipopeptide containing the MPER and the galactosyl ceramide mucosal receptor binding motif, protect non-human primates (NHP) against vaginal heterologous SHIV challenges, in the absence of seric neutralizing antibodies. Correlation was observed between induction of HIV-1 transcytosis-blocking and ADCC activities from cervico-vaginal antibodies and protection. We have then investigated if mucosal antibodies with similar antiviral properties could be induced in women during a Phase I trial.

Background: Macaques vaccinated by intramuscular electroporation with optimized plasmid DNAs expressing SIVmac239 antigens develop potent immune responses able to reduce viremia of the high dose SIVmac251 challenge. To further improve immune responses and protection, protocols combining DNA with protein immunization (coimmunization, prime-boost) were tested. Methods: Indian rhesus macaques (N=8/group) were vaccinated using IM-electroporation by (a) SIVmac239 DNAs only, (b) coimmunization of DNAs with protein (AT-2-inactivated particles) or (c) 2 DNA vaccinations followed by 2 protein boosts (weeks 0, 8, 16 and 36). The animals were challenged weekly with low dose heterologous SIVsmE660 7 months after the last vaccination. Results: Co-delivery of SIV DNA and protein increased the magnitude, longevity, and avidity of humoral responses and the ability to cross-neutralize heterologous Envs. SIV-specific cellular responses were readily measured in blood and mucosal sites after the first immunization and remained high during the entire follow-up. Vaccinated macaques resisted infection by SIVsmE660 compared to nave controls (p=0.05; stratified for Trim5a genotype). Two of 8 DNA-protein coimmunized animals did not get infected after 14 exposures, while all controls were infected by 6 exposures. Vaccinees had significantly lower peak VL (1.7 log, p=0.03) and 75% of vaccinees suppressed virus replication rapidly to undetectable levels, maintaining normal CD4 counts (40 weeks of follow-up). Virus acquisition correlated with antibody avidity to SIVsmE660; acquisition delay and control of viremia also correlated with the presence of vaccineinduced cellular immune responses (CD4+EffectorMemory T-cells and IFN+GranzymeB+cytotoxic T-cells). Conclusion: The vaccine combining DNA and protein in a single administration is a novel concept to achieve rapid, high, persistent, broad and effective immune responses, and captures the best properties of both procedures: high and broad range of cellular immune responses produced by DNA and strong antibody responses produced by protein immunization. This combination is able to prevent infection at the portal of entry.

ORAL ABSTRACT SESSIONS

Methods: This is a double-blind, placebo-controlled Phase I study conducted at CEVAC (Belgium), involving 24 healthy women randomized in 2 Panels to monitor safety, tolerability and immunogenicity of the vaccine MYM-V101 (virosomes-lipopeptides P1): Panel 1: 10 ug/dose and Panel 2: 50 ug/dose. In each Panel, 8 subjects received the vaccine and 4 subjects received the placebo (virosomes without HIV-1 antigens) through intra-muscular (weeks 0/8) and intra-nasal (weeks 16/24) administrations. Results: The vaccine MYM-V101 is considered safe and welltolerated for both Panels, when injected intramuscularly and intransally. All vaccinated subjects have rapidly developed within two months lipopeptide specific serum antibodies. The presence of lipopeptide-P1 specific mucosal IgG and IgA antibodies in the vaginal and rectal secretions was also confirmed for the majority of the samples tested (ImperacerTM technology). Tested vaginal antibodies from the lipopepetide-P1 vaccinated subjects exhibited strong inhibition of HIV-1 transcytosis, as compared to no or weak inhibition with the placebo, confirming previous results from NHP. However, no significant neutralizing activities could be detected in the mucosal samples. We also confirm that MYM-V101 dont induce a Th1 response. Conclusion: This study confirms the safety profile of virosomes and the promising anti-HIV-1 mucosal responses elicited by gp41-derived virosomal vaccine.

80

AIDS Vaccine 2011

Oral Abstract 10: B Cell Vaccine Concepts OA10.05


Direct Antibody Access to the HIV-1 MPER Positively Correlates with Neutralization Sensitivity
B.K. Chakrabarti1, L.M. Walker1, J.F. Guenaga2, A. Ghobbeh2, P. Poignard2, D.R. Burton1, R.T. Wyatt1
1

Oral Abstract Sessions


OA10.06
A DNA Prime/Protein Boost Vaccine Leads to Higher B-Cell Responses than a Vector Prime/ Protein Boost or DNA Prime/Vector Boost Regimens
N. Frahm1, D.P. Friedrich1, P. Walsh1, S. De Rosa1, P. Spearman2, S. Barnett3, M. Keefer4, P. Goepfert5, H. Robinson6, B. Graham7, H. Liao8, G. Tomaras8, B. Haynes8, S. Rerks-Ngarm9, J. Kim10, M.J. McElrath1 Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Emory University, Atlanta, GA, USA; 3Novartis Vaccines and Diagnostics, Cambridge, MA, USA; 4University of Rochester, Rochester, NY, USA; 5University of Alabama at Birmingham, Birmingham, AL, USA; 6Geovax, Atlanta, GA, USA; 7Vaccine Research Center, Bethesda, MD, USA; 8Duke University, Durham, NC, USA; 9Ministry of Public Health, Nonthaburi, Thailand; 10US Military HIV Research Program, WRAIR, Rockville, MD, USA
1 2

International AIDS Vaccine Initiative/The Scripps Research Institute, La Jolla, CA, USA; 2International AIDS Vaccine Initiative, La Jolla, CA, USA
Background: The precise kinetics of neutralization by the gp41 membrane proximal external region (MPER)-directed broadly neutralizing antibodies, 2F5 and 4E10, remains unresolved. Binding data to cell-surface Env and entry data using primary isolates suggests inaccessibility of the 2F5 and 4E10 epitopes on the viral spike prior to receptor engagement, but trimer gel shift analysis and recent slow kinetic shedding data indicate otherwise. Therefore, it remains unclear if the epitopes themselves are formed (or at least sampled) prior to receptor/co-receptor engagement, or if receptor interactions both form and expose the 2F5 and 4E10 epitopes, presumably in the putative pre-fusion transitional intermediate. Methods: Here, we performed antibody-virus washout experiments using both lab-adapted and a diverse panel of clade B primary isolates and one clade C isolate to analyze MPER accessibility. Results: The neutralization activity of 2F5 and 4E10 against labadapted viruses and a few sensitive and moderately resistant viruses was largely unaffected by relatively rapid antibodyvirus washing, suggesting direct interaction with the static spike. However, for more neutralization resistant viruses, the 2F5 and 4E10 antibodies could neutralize only in the no antibody-virus wash conditions, implying that the MPER epitopes were not accessible prior to receptor engagement on these isolates. In fact, accessibility in the washout conditions could be precisely predicted by the relative resistance to neutralization in a standard neutralization format. Conclusion: These data are consistent with a model in which the local MPER antibody epitope conformations may be sampled on the native spike, but are occluded to antibody by local steric or distal quaternary constraints adopted by highly resistant HIV-1 isolates.

Background: Reduced risk of HIV acquisition in RV144 vaccinees in the absence of strong T-cell mediated immunity suggests that protection may be linked to HIV-specific antibodies. We assessed memory B-cell responses to Envelope proteins in RV144 and compared these to responses elicited by other vaccines. Methods: Memory B-cell responses were measured by ELISpot using Env proteins matched to the RV144 protein boosts (gp120_A244, gp120_MN), a consensus (gp140_CON-S) and an unrelated clade B protein (gp140_BaL). Samples from HVTN 049 (3xDNA+2xprotein, n=20), 055 (5xMVA, n=14), 065 (3xMVA, n=20 or 2xDNA+2xMVA, n=20), 068 (2xAd5, n=13) and 204 (3xDNA+1xAd5, n=19) were compared to RV144 (2xALVAC+2xALVAC/2xprotein, n=40); samples obtained at baseline and 2-4 weeks after the last boost of each trial were analyzed. Responses are reported as %Env-specific IgG-producing B cells using total IgG-producing B cells as denominator. No false-positive responses were detected at baseline using 3xbackground (KLH) and >20 spot-forming cells/ million as threshold for positivity. Results: gp140_CON-S responses were detected in 98% of participants post-boost, but magnitudes varied significantly between trials (median 0.18%3.65% Env-specific IgG-producing B cells), with MVA-vectored trials showing low and HVTN049 the highest magnitude. RV144 responses were intermediate, being significantly lower than HVTN049 (p<0.0001) but higher than HVTN055 (p<0.0001). Ad5-vectored vaccines induced responses of similar (HVTN204) or significantly higher (HVTN068, p=0.008) magnitude than RV144. Responses in HVTN049 were stronger than in all other trials (p<0.0001 except HVTN068, p=0.09). Response rates were lower for other proteins but magnitude patterns were similar to those for gp140_CON-S, with RV144 showing a significantly lower magnitude than HVTN049 even for clade AE gp120_A244 (p=0.003). Conclusion: Although strong antibody responses were observed in RV144, memory B-cell responses in this trial are not outstanding in frequency or magnitude compared to other Env-containing regimens, while the clade B DNA prime/protein boost vaccine in HVTN049 generated strong cross-clade memory B-cell responses.

AIDS Vaccine 2011

81

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA11.01
Microarray Analysis of Cervical Tissue Reveals a Transcriptional Imprint that Correlates with Protective Immunity in SIV-Nef-Vaccinated Animals
A.J. Smith1, S.W. Wietgrefe1, C.S. Reilly1, L. Duan1, Q. Li2, M. Zeng1, A.T. Haase1
1

Oral Abstract Session 11: Mucosal Immunity OA11.02


Gut Inflammation and Indoleamine Deoxygenase Inhibit IL-17 Production and Induce Cytotoxicity in RORt+ NKp44+ Mucosal NK Cells During SIV Infection
R. Reeves1, P. Rajakumar1, T.I. Evans1, M. Connole1, J. Gillis1, Y. Kuzmichev1, R. Johnson1
1

University of Minnesota, Minneapolis, MN, USA; 2University of Nebraska, Lincoln, NE, USA

New England Primate Research Center, Harvard Medical School, Southborough, MA, USA

Background: The long-term goal for an efficacious HIV vaccine is to provide sterilizing protection from HIV infection. Thus far, this scenario has only been achieved experimentally using liveattenuated SIV vaccines. As such, a great deal of interest lies in identifying correlates of protection from a successful host response to pathogenic SIV. To this end, we have used a global genomics approach to examine gene expression in the female reproductive tract (FRT) of animals exposed intra-vaginally to WT virus (SIVmac251) and compared this environment to animals first vaccinated with the live-attenuated virus known as SIV Nef and then challenged intra-vaginally with SIVmac251. Methods: RNA from cervical tissue was purified for microarray analysis. The RMA algorithm was used to quantify signal levels for all probe sets and weighted least squares was used to test for differences between groups. Genes were selected based on associated q-values and then functionally classified/annotated while protein expression determined using single-cell analytical procedures for tissue sections. Results: Three major themes emerged from this analysis differentiating the response of vaccinated from unvaccinated animals during virus infection: 1) dampened innate immunity through decreased expression of innate sensors (ex.: MDA5) and increased expression of innate-signaling inhibitors (ex.: PCBP2, ATF7); 2) retarded inflammatory cell infiltration into the FRT of vaccinated animals, coinciding with decreased expression of chemotactic signals (ex.: MIP-1, 3) and increased expression of inhibitors of pro-inflammatory signaling (ex.: RORA, SPRED1, COMMD10); 3) preponderance of gene expression associated with antibody diversification, maturation, and production (ex.:MALT1, IGIP, POLH, ITPKB, RAD52), coinciding with increased plasma cell concentration and immunoglobulin levels in the FRT. Conclusion: This analysis provides both a comprehensive picture of a protective environment in the FRT and tantalizing clues regarding properties of a successful host response to immunodeficiency virus infections (ex.: on-site generation/ diversification of antibody responses and dampened inflammation associated with innate signaling).

Background: Lentiviruses primarily replicate in gut mucosa and cause inflammation and loss of epithelial integrity. Recent studies have identified a novel mucosal NK cell population secreting the TH17 cytokines, IL-17 and IL-22, which are critical for gut homeostasis. However, the effects of lentivirus infection on these unique NK cells are unknown. Methods: Mucosal NK cells, from naive and SIV-infected macaques, were enumerated by a novel bead-based flow cytometry assay, analyzed phenotypically by polychromatic flow cytometry, and evaluated functionally by ICS. Expression analyses of transcription factors and cytokine mRNA in sorted NK cells and whole biopsy pieces were performed by RT-PCR. Results: We identified two lineages of NK cells characterized by dichotomous expression of the NK cell-related markers, NKG2A and NKp44. Both cell types expressed the NK cellrelated transcription factor, NFIL3, but NKp44+ NK cells also expressed the TH17 transcription factor, RORt. Unlike systemic classical NKG2A+ NK cells, NKp44+ NK cells were mucosaerestricted, produced IL-17 and IL-22, but were noncytotoxic. During SIV infection, NKp44+ NK cells were depleted ~9-fold and had decreased IL-17 secretion, increased IFN- secretion, and surprisingly acquired cytotoxicity. NKp44+ NK cells showed no evidence of direct SIV infection in vivo, but depletion and altered function of NKp44+ NK cells were associated with SIV-induced upregulation of inflammatory mediators in the gut, including IDO-1. Furthermore, treatment of NKp44+ NK cells with IDO-1 catabolites in vitro ablated IL-17 production. Conclusion: SIV infection results in massive depletion of NKp44+ NK cells in the gut and reshapes their functional repertoire, not by direct infection, but by indirectly altering the gut cytokine milieu. Due to the critical role of IL-17 and IL-22 in maintaining gut homeostasis, the loss of the NKp44+ NK cell subpopulation is likely to contribute to loss of gut integrity and subsequent microbial translocation and chronic immune activation in lentivirus infections.

ORAL ABSTRACT SESSIONS

82

AIDS Vaccine 2011

Oral Abstract Session 11: Mucosal Immunity OA11.03


Targeting the Vaginal Mucosa with Human Papillomavirus Pseudovirion Vaccines Delivering SIV DNA
S.N. Gordon1, R.C. Kines2, G. Kutsyna1, Z. Ma3, A. Hryniewicz1, B.F. Keele4, J.N. Roberts5, C.Fenizia1, R.Hidajat6, N.Cuburu2, C.B. Buck2, M.L. Bernardo2, M. Robert-Guroff6, C.J. Miller3, B.S. Graham7, D.R. Lowy2, J.T. Schiller2, G. Franchini1
1 National Cancer Institute, National Institute of Health, Bethesda, MD, USA; 2Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, MD, USA; 3California National Primate Research Center, University of California, Davis, CA, USA; 4AIDS and Cancer Virus Program SAICNational Cancer Institute-Frederick, Frederick, MD, USA; 5 Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, MD, USA; 6Immune Biology Retroviral Infection Section, National Cancer Institute, Bethesda, MD, USA; 7 Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA

Oral Abstract Sessions

OA11.04
Durability and Phenotype of SIV-Specific Mucosal T Lymphocyte Responses Elicited by Recombinant Adenovirus Vectors in Rhesus Monkeys
H. Li1, J. Liu1, A. Carville2, K.G. Mansfield2, S. Clark1, D. Lynch1, D.H. Barouch3
1

Beth Israel Deaconess Medical Center, Boston, MA, USA; 2New England Primate Research Center, Southborough, MA, USA; 3 Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
Background: The induction of durable and potent mucosal cellular immune responses may be critical for the development of effective vaccines against HIV-1 and other pathogens. A variety of vaccine vectors have been reported to elicit HIV-1/ SIV-specific cellular immune responses at mucosal sites, but the anatomic distribution, kinetics, durability and phenotype of these responses remain poorly characterized. Methods: Rhesus monkeys were immunized with intramuscular injection of (i) single, (ii) homologous prime-boost, or (iii) heterologous prime-boost regimen with recombinant adenovirus (rAd) vectors expressing SIVmac239 Gag. Magnitude and phenotype of SIV-specific T lymphocytes were evaluated with multiparameter tetramer staining assays and multiparameter intracellular cytokine staining (ICS) assays. Results: Intramuscular injection of rAd vectors in rhesus monkeys induced durable SIV-specific T lymphocyte responses that persisted for over 2 years in multiple mucosal tissues, including colorectal, duodenal and vaginal biopsies as well as bronchoalveolar lavage (BAL). In peripheral blood, SIV-specific T lymphocytes underwent a phenotypic evolution from effector memory (EM) and transitional memory (TM) cells to central memory (CM) cells following vaccination. In contrast, mucosal SIV-specific T lymphocytes exhibited a persistent TM phenotype as well as persistent activation characterized by CD69 expression. Conclusion: These data demonstrate that rAd vectors induce durable and widely distributed mucosal T lymphocyte responses that are phenotypically distinct from peripheral T lymphocyte responses. These findings suggest that vaccine-elicited T lymphocytes acquire qualitative features of target tissues following trafficking to mucosal sites which may allow them to respond rapidly upon mucosal viral challenge.

Background: Most HIV transmission occurs by the genital route, suggesting that vaccines that target the genital mucosa and induce local B- and T-cell immune responses may offer better protection from infection than vaccines that induce mainly systemic responses. Methods: We tested a novel vaccine delivery platform that specifically targets the vaginal mucosa. Human Papillomavirus (HPV) capsid proteins, L1 and L2, can self assemble into virus like particles (VLPs), and when co-transfected with a plasmid containing a gene of interest, L1 and L2 will encapsidate the gene and form pseudovirions (PsV). We generated HPV PsVs vaccines that deliver SIV genes to the female genital tract and designed feasibility and vaccine protection studies, using the SIVmac251 macaque model. Results: In the feasibility study, an HPV PsV Gag vaccine induced SIV-specific IgG and IgA in vaginal secretions and SIVspecific T-cell responses in the cervicovaginal tract of macaques. Importantly, HPV PsV- induced mucosal T-cell responses that rapidly expand upon vaginal exposure of a subset of animals to high doses of SIVmac251. To evaluate relative vaccine efficacy,a cohort of macaques have been immunized using HPV PsVs, delivering SIV Gag-pro, and env genes, as well as a re-assorted gene encompassing the Tat, Nef, and Rev genes (Retanef). The animals were then boosted, with recombinant gp120 protein. The efficacy of a systemic prime and mucosal boost vaccine regimen is also being evaluated by combining the canarypox vector ALVAC and HPV-PsV. At the end of the immunization regimen (June 2011), these animals will undergo repeated lowdose intravaginal challenges with SIVmac251 to mimic HIV transmission among humans. Conclusion: The HPV PsV system is a novel approach for direct genetic vaccination of the vaginal mucosa and is immunogenic in macaques. SIV challenge data to determine whether this combination of vaccine modalities will afford significant protection from SIVmac251 infection will be presented.

AIDS Vaccine 2011

83

ORAL ABSTRACT SESSIONS

Oral Abstract Sessions


OA11.05
Role of Genital Tract Inflammation and T Cell Activation in CD4 Depletion at the Female Genital Tract During HIV Infection
L. Liebenberg1, H. Jaspan2, D. Coetzee3, A. Williamson1, J. Passmore1
2

Oral Abstract Session 11: Mucosal Immunity OA11.06 LB


SIV Attenuated Vaccine-Induced Plasma Cells and Antibodies Limit Infection at the Portal of Entry and Systemically Following Vaginal Challenge
M. Zeng1, L. Duan1, Q. Li2, A. Smith1, S. Wietgrefe1, P.j. Southern1, C.S. Reilly1, S. Pambuccian1, L. Shang1, P.J. Skinner1, M. Zupancic1, M. Piatak, Jr3, D. Waterman4, K. Reeves5, J.D. Lifson3, P. Johnson6, A.T. Haase1
1 University of Minnesota, Minneapolis, MN, USA; 2University of Nebraska, Lincoln, NE, USA; 3National Cancer Institute, Frederick, MD, USA; 4ZeptoMetrix Corporation, Buffalo, NY, USA; 5Harvard Medical School, Southborough, MA, USA; 6Harvard Medical School and Ragon Institute, Southborough, MA, USA

University of Cape Town, Cape Town, South Africa; Department of Pediatrics, Seattle Childrens Hospital, University of Washington, Seattle, WA, USA; 3School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
1

Background: The micro-environment within the female genital tract has a strong influence on both the acquisition and transmission of HIV-1 during penetrative sex. Although immune activation in response to invading pathogens is a crucial to protective immunity, such responses may ironically also contribute to HIV pathogenesis by providing the virus with a steady supply of susceptible target cells. This study investigated the role of inflammation and immune activation in CD4 depletion and HIV shedding in the female genital tract associated with HIV pathogenesis. Methods: Cervical cytobrush and blood-derived CD8+ and CD4+ T cells from 33 HIV+ and 40 HIV- women were investigated for expression of activation markers CCR5, HLA-DR, CD38 and Ki67 by flow cytometry. Concentrations of inflammatory and regulatory cytokines (eotaxin, G-CSF, IL-1, IP-10, MIP-1, IL6, IL-10, IL-8 and RANTES) in cervical supernatants and blood plasma were determined by Luminex. HIV loads were measured in cervical secretions and plasma. Results: The frequency of activation marker expression in blood was significantly predictive of genital mucosal T cell activation in HIV-infected women. Importantly, cervical T cell activation was broadly associated with mucosal HIV shedding and the extent of CD4 depletion in the genital tract during HIV infection. HIV-infected women displayed greater levels of T cell immune activation in the blood and cervix than uninfected women, and had significantly elevated frequencies of activation in the cervix than in blood. IL-8 and G-CSF levels were consistently greater at the cervix than blood. Genital tract concentrations of the inflammatory chemokine RANTES correlated positively with local CD38+ and HLA-DR+ T cell frequencies, with local HIV shedding, and negatively with CD4 depletion. Conclusion: We conclude that the interplay between mucosal inflammation and local immune activation is an important contributor to CD4 depletion and the degree of HIV shedding in the female genital tract during HIV infection.

Background: In the SIV rhesus macaque model of HIV-1 transmission to women, vaccination with SIVmac239-delta-nef confers partial or complete protection against vaginal challenge with SIVmac251 by as yet unknown mechanisms. Methods: To identify potential protective mechanisms against vaginal transmission, we analyzed host responses in cervicalvaginal and lymphatic tissues (LTs) of Indian rhesus macaques following intravenous SIV delta-nef infection (vaccine phase), and following vaginal challenge with SIVmac251 (Challenge phase). Results: We found that vaccine induces a significant increase in SIV-specific antibodies and plasma cells and IgG localized beneath the cervical-vaginal epithelium and the development of ectopic lymphoid tissue follicles within cervical-vaginal tissues. Within the first week after heterologous vaginal challenge of SIVmac251, there is a rapid increase in the number of IgG producing plasma cells, concentration of anti-Env antibody and the breadth of antibody in vaginal-cervical mucosal sites in the vaccinated rhesus macaques, which correlates with the local expansion and maturation of ectopic lymphoid follicles within vaginal tissues in which there are plasmablasts and/or memory B cells with evidence of AID-mediated class-switch recombination. The high concentrations of antibodies and plasma cells correlate with as much as a 6-order of magnitude reduction of tissue viral load at the portal of entry. There is a similar rapid increase in plasma cells in the lymphoid tissues that correlates with a rapid response to disseminated infection that is also associated with major reductions in viral replication at these sites. Conclusion: These data suggest a mechanism by which a humoral response at the right place, right time may contribute to the protection, a concept relevant to developing effective vaccines against HIV-1 and other mucosal pathogens.

ORAL ABSTRACT SESSIONS

84

AIDS Vaccine 2011

Posters
Acute HIV Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 87 Adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 91 Animal Models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 96 B Cell Immunology and Neutralizing Antibodies . . . . . . . . pg 97 B Cell Vaccine Concepts and Design . . . . . . . . . . . . . . . . . . pg 118 Clinical Trial Site Challenges . . . . . . . . . . . . . . . . . . . . . . . . . pg 122 Host Genetic Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 130 Immune Escape . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 133 Innate Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 135 Mucosal Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 143 Novel Immunogens/Inserts . . . . . . . . . . . . . . . . . . . . . . . . . . pg 148 Pediatric/Adolescent Infections and Trials . . . . . . . . . . . . . . pg 152 Preclinical and Clinical Therapeutic Trials . . . . . . . . . . . . . . pg 154 Preclinical and Clinical Prophylactic Vaccine Trials . . . . . . pg 155 Prevention Strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 162 Social/Ethical/Access/Regulatory Issues . . . . . . . . . . . . . . . . pg 169 T Cell Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 174 T Cell Vaccine Concepts and Design . . . . . . . . . . . . . . . . . . . pg 191 Vaccine Concepts and Design . . . . . . . . . . . . . . . . . . . . . . . . pg 205 Viral Diversity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pg 222 Virology and Immunology of HIV Transmission . . . . . . . . . pg 227

AIDS Vaccine 2011

85

POSTERS

86

AIDS Vaccine 2011

Topic 01: Acute HIV Infection P01.01


Analysis of Epitope-Specific HIV T Cell Responses During Early HIV-1 Infection and Their Association with Viral Control
P. Kunwar , D. He , N.R. Hawkins , Y. Liu , A. Collier , T. Hertz , P. Gilbert3, H. Horton7
1 2 3 4 5 6

Posters
P01.02
Balance of CTL Escape and Viral Fitness in Acute and Early HIV Infection
J. Brown1, M. Rolland2, J.T. Herbeck 1, N. Frahm3, J.I. Mullins1 University of Washington, Seattle, WA, USA; 2U.S. Military HIV Research Program, Henry M. Jackson Foundation, Rockville, MD, USA; 3Fred Hutchinson Cancer Research Center, Seattle, WA, USA
1

University of Washington, Seattle, WA, USA; 2University of California Los Angeles, Los Angeles, CA, USA; 3Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Seattle, WA, USA; 4Department of Microbiology, University of Washington, Seattle, WA, USA; 5Department of Medicine, University of Washington, Seattle, WA, USA; 6Vaccine and Infectious Disease Division, Fred Hutch Cancer Research Center, Seattle, WA, USA; 7University of Washington and Seattle Biomedical Research Institute, Seattle, WA, USA
1

Background: Investigations into host-virus interactions during acute HIV-1 infection are important for the design of a preventative HIV-1 vaccine. To understand the dynamic interplay between host CTL responses and the mechanisms by which HIV-1 evades those responses, we studied viral evolutionary patterns and host CTL responses in a linked transmitter:recipient pair. Methods: Ten HIV-1 SGA sequences were amplified from the transmitter T3 near the time of transmission and from the recipient R3 at sequential visits from 25-247 days post-onset of symptoms (dps). A composite alignment of all sequences was generated to identify the amino acids (AA) sites that i) differed between T3 and R3 or ii) evolved over time in R3. To identify if CTL pressure promoted these AA changes, IFN ELISpot assays were performed on T3 and R3 using an autologous peptide set designed against the regions harboring evolving sites and including variants found in T3 and R3. Results: We identified responses in R3 against wildtype and variant forms of peptides, sometimes at a time when the variant sequence had not yet been detected, suggesting that crossreactive CTL failed to contain the early evolution of mutant viruses. In R3, CTL-mediated escape mutations were identified in the functionally constrained regions of the Gag-Pol frameshift stimulator loop and in the Env gp41 C-terminal Heptad Repeat. The virus appears to have exploited the limited availability of AA sites not required to maintain function in these regions in order to attain CTL escape. Conclusion: Focusing immunological studies on viral regions under selection pressure allows for detailed investigations of hostvirus interactions using limited numbers of PBMC. Comparison of viruses between the transmitter and recipient provides a better characterization of founder viruses specificities. Identification of escape mechanisms allows further understanding of the challenges faced by the virus in achieving a balance of immunologic escape and replicative fitness costs.

Background: HIV-1-specific cytotoxic T lymphocytes (CTL) are important in the control of viremia; however, the precise qualities of effective epitope-specific T cell responses that may be responsible for control remain unclear. Here we investigated the frequency and specificities of T cell responses during early HIV-1 infection, to address the question of whether conservation score (CS) of targeted epitopes play an important role in viral control. Methods: Using IFN- ELISpot assays, we mapped epitope specificities recognized by HIV-specific T cells in 12 ART-nave individuals during early infection (within 6 months). We then identified CS of targeted epitopes, where the CS is defined as the proportion of HIV-1 group M amino acid sequences in the LANL database that include the epitope. We further evaluated the association between the CS of the targeted epitopes and the individuals viral-load. Results: Our epitope mapping of T cell responses showed that the median number of epitopes targeted was 9 (range, 5-24) mostly against Gag, Nef, Env and Pol. More than one third of these epitopes were novel epitopes. In addition, variable epitopes were targeted more frequently than conserved epitopes (p<0.0001). The median CS of targeted Gag epitopes were more conserved than those of Pol, Nef, Env and Acc epitopes (p=0.003). However, the magnitude of responses elicited by Gag epitopes was not significantly different than those elicited by Pol, Nef, Env and Acc epitopes. Furthermore, there was a significant inverse correlation between the rank of CS of targeted epitopes with the rank of viral-load (r= -0.76, p=0.004), indicating that individuals that possess T cells targeting conserved epitopes early in infection had lower viral loads. Conclusion: These data suggest that HIV-specific CTL targeting conserved epitopes of HIV-1 are superior at controlling viral replication in vivo, supporting the rationale for designing vaccines containing conserved immunogens. This project is funded by NIH grant #R01AI090783.

AIDS Vaccine 2011

87

POSTERS

Posters
P01.03

Topic 01: Acute HIV Infection P01.04


Increased Plasma Levels of Hemoxygenase-1 (HO-1) and Presence of HO-1-Specific Regulatory T Cells in Acute HIV-1 Infection
M. Angin1, A. Fathi1, M. King1, M.M. Addo1
1

Abnormal Liver Function During Acute HIV Infection Correlates with Fiebig Stage and Systemic Inflammation
J. Fletcher1, W. Ridtitid2, M. Marovich3, I. Sereti4, R. Rerknimitr2, A. Schuetz5, S. Pinyakorn1, D. Suttichom1, S. Rattanamanee1, R. Trichavaroj6, V. Nguay6, J.H. Kim6, J. Ananworanich1
1 SEARCH, The Thai Red Cross AIDS Research Centre, Bangkok, Thailand; 2Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; 3US Military HIV Research Program, Rockville, MD, USA; 4National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA; 5Division of Retrovirology, Armed Forces Research Institute of Medical, Bangkok, Thailand; 6Division of Retrovirology, AFRIMS, Bangkok, Thailand

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA

Background: Heme-oxygenase-1 (HO-1) the rate-limiting enzyme in heme catabolism is an inducible stress-response protein with potent anti-inflammatory activity. Recent studies in cancer patients described a novel cell type of HO-1 specific regulatory CD8+ T cells with suppressive capacity superior to conventional Tregs. The role of these cells in HIV-1 infection has not been explored to date; however, if detectable they could represent potent regulators of HIV-1 immunity and hold potential as a novel target for future immuno-therapeutic or vaccine interventions. Methods: Hemoxygenase-1 protein levels were measured in plasma from 40 individuals with untreated primary HIV-1 infection, 39 elite controllers, 39 chronic progressors and 15 healthy controls. The frequency of HO-1 specific CD8+ T cells was measured by flowcytometry using HLA class I pentamers in PBMC from individuals with untreated primary HIV-1 infection, elite controllers, chronic progressors and healthy controls. Results: The mean plasma concentration of heme-oxygenase-1 was 1.7ng/ml in healthy control subjects. HO-1 plasma levels were significantly increased in individuals with acute HIV-1 infection (mean = 3.3ng/ml) compared to HIV uninfected control subjects. HO-1 plasma levels did not correlate with CD4 count or viral load. HO-1-specific CD8+ T cells were detectable in HIV1 infected individuals with the highest levels observed during primary HIV-1 infection. Preliminary data showed evidence for suppressive activity of supernatant from cultured sorted HO-1specific CD8+ Tregs in HIV-1-specific cytotoxicity assays. Conclusion: Our data demonstrate elevated levels of hemeoxygenase-1 in individuals with acute HIV-1 infection suggesting that HO-1 is induced during this phase and likely mediates anti-inflammatory effects. Moreover, the presence of the HO-1 antigen during acute infection appears to drive the elevated frequency of HO-1-specific CD8+ regulatory T cells in acutely infected individuals. The investigation of this novel cell type may provide further insight into HIV-1 T cell regulation via nonvirus specific immune mechanisms and hold potential for future immunotherapeutic intervention.

Background: Abnormal liver function may present during acute HIV infection (AHI). Its pathogenesis is unclear but may be linked to systemic inflammation. Methods: 30 Thai adults with Fiebig stages I to IV AHI were assessed at baseline for ALT, yGT, bilirubin, IP-10, MCP-1, TNF, IL-22, IL-6 (n=30) and markers of inflammation [CRP, d-dimer, hyaluronic acid (n=19), sCD14 (n=7)]. The % CD4+CCR5+ cells in sigmoid tissue was determined by flow cytometry (n=24). Results: Twenty-six (87%) were male; the mean (SD) age was 29 (6.2) years; they were 8 Fiebig I; 5 Fiebig II; 14 Fiebig III and 3 Fiebig IV. Mean (SD) time from HIV exposure was 18 (9.1) days. Median (IQR) CD4 was 406 cells/mm3 (298-555) and HIV RNA was 5.5 (5.1-6.4) log10copies/ml. Twenty-six (87%) had ARS. Most common symptoms were fever (77%), myalgia (60%), fatigue (57%), and headache, pharyngitis, and oral ulcers (all 50%). Three had positive HBsAg and none had positive HCV Ab. Sixteen (53%) had abnormal ALT (>31 UL-1) and 10 (33%) had abnormal yGT (>43 UL-1); 2 of these had positive HBsAg (ALT 45 and yGT 49 respectively). Absolute ALT and yGT positively correlated with Fiebig stage [rho=0.689 (p<0.001) and rho=0.518 (p=0.003) respectively]. Abnormal ALT was more common in Fiebig III/IV compared to Fiebig I/II (OR=13.2, p=0.009). Both absolute and abnormal yGT correlated with TNF- expression (both rho=0.406, p=0.026). Absolute ALT was positively correlated with IP-10 (rho=0.403, p=0.027), while abnormal ALT was correlated with IL-12 (rho=0.375, p=0.041). No relationship between ALT/yGT with gut CD4+CCR5+ T cells or CRP, D-dimer, HA and sCD14 was seen. Conclusion: ARS was common in Thais with AHI. Abnormal liver function was prominent and correlated with Fiebig stage and inflammatory cytokines levels, but not with gut T cell depletion. The clinical and prognostic implications of these findings require further study.

POSTERS 88 AIDS Vaccine 2011

Topic 01: Acute HIV Infection P01.05


Early Cytokine and Chemokine Patterns During Acute HIV Infection
B. Tassaneetrithep1, B. Slike2, S. Nitayaphan3, K. Rono4, E. Sanga5, A. Sekiziyvu6, K. Dambach2, G. Donofrio2, M. Creegan2, J. Kim2, N. Michael2, M. Robb2, M. Marovich2 Faculty of Medicine Siriraj Hospital, Mahidol University and MHRP, Rockville, MD, USA; 2US Military Research Program, USA; 3AFRIMS, Bangkok, Thailand; 4WRP-Kenya, Kericho, Kenya; 5MMRP, Mbeya, United Republic of Tanzania; 6 MUWRP, Kampala, Uganda
1

Posters
P01.06
Whole Blood Transcriptional Monitoring of Acute HIV-1 Infection Reveals Differential Signatures of Host Immune Activation
J.A. Skinner1, N. Baldwin1, M. Sharma1, B. Lemoine1, D. Blankenship3, H. Qin3, Z. Xu1, E.B. DeVol3, V. Pascual1, A. Mejias4, O. Ramilo4, R. Pankla5, G. Lertmemongkolchai6, M. Berry7, A. OGarra7, K. Palucka1, R. Thiebaut8, G. Chene8, M. Cohen9, K. Soderberg10, T.N. Denny10, A. McMichael11, B.F. Haynes10, Y. Levy2, J. Banchereau1, D. Chaussabel1, NIAID Center for HIV/AIDS Vaccine Immunology1 Baylor Institute for Immunology Research, Dallas, TX, USA; Henri Mondor Hospital-ANRS, Creteil, France; 3Baylor Institute for Health Care Research and Improvement, Dallas, TX, USA; 4Nationwide Childrens Hospital Center for Vaccines and Immunity, Columbus, OH, USA; 5Department of Medical Technology, Naresuan University, Phitsanulok, Thailand; 6 Department of Clinical Immunology, Khon Kaen University, Khon Kaen, Thailand; 7MRC National Institute for Medical Research, London, United Kingdom (Great Britain); 8INSERM U897, Bordeaux, France; 9University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; 10Duke University School of Medicine, Durham, NC, USA; 11Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom (Great Britain)
1 2

Background: Understanding the earliest immunologic events during acute HIV infection is critical to inform prevention and treatment efforts. We focused on characterizing plasma cytokine profiles during the earliest stages of acute infections from subjects in the prospective RV217 cohort representing multiple HIV subtype infections (A, C, D and CRF-01). Methods: Twice weekly nucleic acid testing (NAT) was used to prospectively identify subjects during very early HIV infection (NAT+, antibody-). Plasma cytokine levels were measured at baseline and at all available early time points throughout acute infection and early set point. Assays were performed in triplicate using the Quansys Q-Plex Multiplex IR ELISA Array. Images were captured using the LiCor IR imaging system and analyzed using Q-View Plus software. Data and statistical analysis were performed using GraphPad Prism. Results: Longitudinal plasma samples acquired prior to EIA reactivity during the upslope in viral load from acutely infected individuals (n=6) showed similar cytokine profiles. Plasma levels of the CC-chemokine MCP-1/CCL2 (6/6) and CXC-chemokine IP-10/CXCL10 (4/4) rose sharply during viral load ramp up. Conversely, a concomitant nadir in plasma IL-2 (5/6), often with a decrease in IL-17 (4/6) was observed. There were profound decreases in both peripheral absolute CD4 counts and B cells during these early days of viral expansion. Other cytokines were detected (IL-1, IL-6, IL-10, IL-12p70, IL-15 and IFN-) but showed more variability in expression patterns during early phases of infection. Conclusion: Few studies have examined cytokine expression patterns prior to and during the acute phase of HIV infection. The early detection of MCP-1/CCL2 and IP-10/CXCL10 observed here, similar to a prior study in acute subtype B infections, may indicate early trafficking events and reflect involvement of specific cell types during acute infection. Falling levels of IL-2 and IL-17 could be linked to changes in frequency or redistribution of peripheral lymphocyte subsets.

Background: Genome-wide transcriptional studies in nonhuman primates have suggested that the quality and magnitude of early immune responses to SIV infection are associated with downstream disease progression. The aim of this study is to identify, monitor, and interpret the whole blood transcriptional signature of acute HIV-1 infection (AHI). Methods: A total of 56 AHI patients and matching healthy controls from both Africa and the United States were distributed into training, test, and validation datasets. Whole blood from these subjects was obtained for transcriptional profiling using microarrays. Both gene and modular transcriptional framework analyses were utilized to interpret the signature of AHI and for comparisons with other disease signatures including: influenza, tuberculosis, sepsis, and chronic HIV following interruption of anti-retroviral therapy (ART). Additional samples were collected at 1, 2, 4, 12, and 24 weeks post-enrollment for training and test set AHI subjects, to establish the longevity of the signature in the presence or absence therapy. Results: We identified a robust AHI signature with increased activity in: interferon, cell cycle, cytotoxic, plasma cell, and B-cell modules amongst others. This activity was unique when compared to signatures obtained from patients with other infections. Only interferon and cell cycle activity was observed in both AHI subjects and chronically infected patients following interruption of therapy. Notably, 20% of AHI patients were observed to express a quiescent signature that clustered independent of the highly active signature described above. We found that these patients had significantly lower viral set-points (p=0.01) when compared to patients with the active signature. Finally, longitudinal analysis demonstrated that the active AHI signature can persist independent of viral load in ART treated patients and up to 6 months in the absence of therapy. Conclusion: Transcriptional monitoring of early HIV infection offers both quantitative and qualitative assessments patient responses that may be predictive of long-term disease progression.

AIDS Vaccine 2011

89

POSTERS

Posters
P01.07

Topic 01: Acute HIV Infection P01.08


Integrated Analysis of Immunogenicity, Viral Sequence, and Viral Load Data in the Step Study
H. Janes1, P. Gilbert1, N. Fraham1, M. Rolland2, A. DeCamp1, E. Gabriel3, J. Wolfson4, J. Mullins3, J. McElrath1 Fred Hutchinson Cancer Research Center, Seattle, WA, USA; U.S. Military HIV Research Program, Rockville, MD, USA; 3 University of Washington, Seattle, WA, USA; 4University of Minnesota, Minneapolis, MN, USA
1 2

Risk Behavior and Demographic Characteristics of Infected vs Non-Infected Participants in RV217d, a Study in At-Risk Populations in Pattaya, Thailand
A. Bolen Valenzuela , P. Morgan , M. Benenson , S. Sriplienchan2, N. Thaitawat2, P. Buapunth2, K. Pumratana2, J. Kim1, N. Michael1, M. Robb1
1 2 2 1 2

U.S. Military HIV Research Program, Rockville, MD, USA; Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand

Background: RV217d is a study of early capture HIV infection in most at risk populations in Pattaya, Thailand through repeat twice weekly testing. Methods: Female sex workers (FSW), MSM, and transgenders (TG) were given a baseline risk assessment. Those at risk were enrolled and followed for two years with twice weekly HIV testing. Results: A total of 381 participants took the baseline questionnaire. The majority reported working as bar, massage or sex workers (94%) or having received money in exchange for sex (85%). They are well educated, with average age of 27 years and 39% have never married. At baseline their recent risk for HIV is high, with unprotected sex with a known HIV positive partner, 15%; unprotected sex with greater than or equal to three partners in the last three months, 43%; recent STI symptoms, 60% and inconsistent or no condom use with male clients, 56%. Sixteen participants have become infected to date, for an incidence rate of 8.59/ 100 py in MSM, 6.81 in TGs, and 2.10 in FSWs. Incident cases are younger, less educated, and earn half the income of the uninfected group. The groups were similar across the behavioral responses except 77% of infected MSM and TG participants had sex exclusively with males or transgenders versus 59% of the uninfected. Conclusion: In this most at risk cohort, a lack of a characteristic behavioral profile distinguishing infected participants may indicate that all are at risk; however, low-income, younger MSM and TGs would be an appropriate group to target.

Background: The sieve analysis for the Step Study (Rolland et al. 2011) showed that breakthrough HIV-1 sequences were more divergent from the vaccine insert in vaccine than placebo recipients, and identified 10 signature sites where the rate of amino acid (AA) mismatch to the insert residue differed between treatment groups. We linked Step viral sequences with immunogenicity and acute viral load data to evaluate mechanisms for, and consequences of, the sieve effect. Methods: Analyses included 91 male subjects diagnosed with HIV infection prior to study unblinding (37 placebo, 54 vaccine recipients). Viral sequences from the first PCR-positive visit were summarized using global measures of genetic distance and the 10 AA signature sites identified by Rolland et al. Pre-infection T-cell responses were mapped to individual 15-mers 4 weeks post second vaccination using interferon-gamma (IFN) ELISpot. Acute viral load, obtained at RNA-positive but antibody-negative visits, was available for 27 subjects and multiply imputed for the remainder. Results: There was some evidence of reduced acute viremia in vaccines versus placebos (4.7 vs 5.1 log RNA copies/ml), although this difference was not significant (p = 0.27) and not predicted by pre-infection T-cell responses. The locations of the epitopes targeted pre-infection did not match the signature sites. Magnitude and breadth of the pre-infection responses were not associated with global genetic distances. Vaccine and placebo recipients did not differ in their associations between global genetic distance and acute viral load; however, mutations at five Gag AA sites were associated with acute viral load in vaccinees. Conclusion: The Step vaccine not only impacted founder virus populations, but potentially reduced acute viral load. However, current measures of T-cell function did not predict these effects. Sequence differences between treatment groups were not associated with viral load, although we did identify several mutations in Gag associated with lower/higher viral load in vaccinees alone.

POSTERS 90 AIDS Vaccine 2011

Topic 02: Adjuvants P02.01


Specific IgA Plasma Cell Recruitment to the Female Genital Tract Following Intravaginal Administration of CCL28 or MPLA
V. Buffa1
1

Posters
P02.02
Vaccine Antigen Immunity Is Enhanced by Optimized TLR4 and TLR7/8 Adjuvant Combinations in Mini-Pigs Inoculated via a Systemic but Not a Mucosal Route
P.F. McKay1, D.F. King2, D. Carter3, R.J. Shattock2
1

Imperial College, London, United Kingdom (Great Britain)

Background: Priming of immune response by the NALT is known to induce responses in the female genital tract and the rectum. This is in line with the concept of a common mucosal immune system whereby a fraction of B cells primed at one mucosal site can re-circulate through the systemic compartment and repopulate distant mucosal compartments. CCL28 is thought to be central to recruitment of IgA plasma cells to the genital tract and colorectum. In addition, inflammation, such as that induced by TLR activation, is also associated with plasma cell recruitment. In this study we assess whether vaginal application of CCL28 or TLR4 ligand MPLA, enhanced immune response following intranasal or parenteral immunization. Methods: Animal (n=8 mice/group) received three intranasal (IN) or subcutaneous (SC) immunizations with gp140 (10ug) plus MPLA (20ug). Six days following each immunization CCL28 (10ug), MPLA (20ug) or PBS, was applied to the vagina. Blood and vaginal samples were taken to detect specific IgG and IgA responses. Vaginal and spleen cells were isolated to perform B and T cell assays. Results: IN immunization induced systemic and vaginal specific IgG and IgA responses. In contrast SC immunization induced lower vaginal specific IgG responses and no vaginal IgA. Intravaginal administration of either CCL28 or MPLA six days post IN immunization significantly enhanced vaginal antibody responses, MPLA providing greater enhancement. Intra-vaginal MPLA administration also enhanced systemic IgA levels and produced detectable vaginal cellular responses. The impact of CCL28 or MPLA on SC immunization was minimal. Conclusion: This study shows that specific B cell responses following intranasal immunization can be enhanced by local application of MPLA or CCL28 in the vaginal six days post immunization. Exploitation of the mechanisms leading to enhanced responses at mucosal surfaces could be an important component in the design of protective vaccines.

St Georges, University of London, London, United Kingdom (Great Britain); 2Imperial College London, London, United Kingdom (Great Britain); 3Infectious Disease Research Institute, Seattle, WA, USA

Background: Previous studies have demonstrated synergy between TLR4 and TLR7/8 stimulation with enhanced CD4 T cell cytokine production and cooperative upregulation. We performed a series of experiments to analyse the inter-relationship of these two innate receptors in mini-pigs, animals that are fully responsive to these TLR ligands and that are immunologically similar to man. Methods: Groups of 4 pigs were vaccinated twice at monthly intervals with protein antigens adjuvanted with various concentrations of either aqueous formulation GLA (synthetic monophosphoryl lipid A - TLR4 ligand) or R848 (resiquimod - TLR7/8 ligand) via the intranasal (IN - 50ug antigen) or intradermal (ID - 20ug antigen) route. Pigs were sampled weekly (serum and vaginal wash) and antigen-specific IgG and IgA quantified by ELISA. Results: R848 augmented antigen-specific responses when administered via the IN or ID route, while an adjuvant effect was only observed with GLA used ID. Combinations of optimal amounts of each TLR ligand had an additive enhancing effect when used ID but surprisingly GLA decreased R848-induced immunity after IN inoculation. Interestingly, R848 significantly enhanced mucosal antigen-specific IgA after IN inoculation but only within a narrow concentration range, as high dose R848 inhibited mucosal IgA. Conclusion: These data begin to address important issues relating to adjuvant combinations and route of administration. TLR4 and TLR7/8 agonists combined to additively enhance antigen-driven immune responses but only after ID vaccination. The inhibitory effect of GLA on R848-driven responses suggests an active contribution by GLA rather than a simple formulation issue, as adjuvant co-formulation did not inhibit R848 responses after ID vaccination. While high dose R848 did not inhibit the serum antibody responses we found that mucosal responses, particularly IgA, were particularly dose sensitive and responses were significantly abrogated above a threshold, a crucial parameter to incorporate into any adjuvanted vaccine modality.

AIDS Vaccine 2011

91

POSTERS

Posters
P02.03

Topic 02: Adjuvants P02.04


A Novel Glycolipid Adjuvant Significantly Boosts Cellular Immune Responses to an Adenoviral Vaccine in Non Human Primates
S. Vasan1, M. Boente-Carrera1, J. McManus1, N.N. Padte1, A. Gettie1, B.F. Grasperge2, N. Patterson3, C.D. Andrews1, T. Richie4, M. Tsuji1 Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY, USA; 2Tulane National Primate Research Center, Covington, LA, USA; 3Henry M. Jackson Foundation, Rockville, MD, USA; 4U.S. Naval Medical Research Center, Silver Spring, MD, USA
1

A Comparative Analysis of HIV-Specific Immunity Following DNA Co-Immunisation with Different Adjuvants
J. Hou1
1

Chinese Center for Disease Control and Prevention, Beijing, China

Background: In this research, we investigated the efficacy of combined use of each adjuvant such as CpG, BCG-PSN (Polysaccharide nucleic acid fraction from BCG), flagellin, PA-MSHA (Pseudomonas aeruginosa mannose-sensitive hemagglutination pilus strain), CTB(Cholera toxin B subunit) with DNA vaccine in comparison to DNA vaccine alone in inducing immunity against HIV-1, to improve DNA vaccine immunogenicity. Methods: In this study, 6-8-week-old female BALB/C mice were administrated intramuscularly with different dosages (10g, 50g) pDRVI1.0-gp1455m HIV-1 DNA vaccine co-formulated with each adjuvant or DNA alone at 3 weeks interval. IFN- ELISPOT, envspecific ELISA, IgG isotype ELISA and avidity ELISA detected cellular and humoral immunological response, respectively. In vitro, we stimulated bone marrow-derived dendritic cells (BMDC) with adjuvants, and analyzed activation of DCs by measuring expression of surface molecules. Results: HIV DNA co-immunization with five adjuvants induced more robust immune responses. BCG-PSN, PA-MSHA, CTB generated an average of 250 SFCs/million, 280 SFCs/million and 330 SFCs/million, co-inoculated with DNA, respectively, whereas DNA alone generated an average of 180 SFCs/million. However humoral response showed no difference between DNA co-inoculation with adjuvant (average GMT=12800) and DNA alone significantly. We found out that vaccination with 10g DNA co-formulation adjuvants (170-200SFC/million) induced similar immune response compared with 50g DNA alone (180SFC/million). In the avidity experiment, results demonstrated that almost all adjuvants especially CTB (ED50=1.8) promoted production of high-affinity antibody. In vitro, adjuvants stimulated BMDC indicated several molecular such as CD40 (MFI, range 394 to 3537 versus 294), CD80 (MFI, range 468 to 900 versus 456) and CD86 (MFI, range 697 to 1660 versus 385) up-regulation, comparison with negative control, it meant that adjuvants activated DCs, especially PA-MSHA and flagellin showed powerful DCs activators. Conclusion: CpG, BCG-PSN, flagellin, PA-MSHA and CTB as potent adjuvants can enhance the quality of immune response especially promoting antibody avidity and DCs activation.

Background: The majority of adjuvants in development to date boost immune responses to protein antigens. Identification of effective adjuvants for viral vectors remains critical to improving vaccination regimens that utilize viral vectors. We have identified a novel glycolipid compound, 7DW8-5, which binds the CD1d receptor with higher affinity than its parent compound, -galactosyl ceramide (GalCer), enabling NKT cell activation and subsequent recruitment and activation of dendritic cells and other inflammatory mediators. We sought to determine whether 7DW8-5 would provide an adjuvant effect to an adenoviral vaccine in rhesus macaques, and to determine the optimal dose for subsequent clinical development. Methods: Five groups of rhesus macaques (n=5 per group) received a single intramuscular vaccination with 2x1010pu NMRCM3V-Ad-PfCA (AdPfCA), a 1:1 mixture of Ad5-based vectors expressing the CSP and AMA-1 antigens from Plasmodium falciparum, respectively, with increasing concentrations of 7DW8-5 in each group: no adjuvant, 0.1g, 1g, 10g, and 100g. Macaques were monitored for safety, innate immune responses and immunogenicity for four weeks. Results: 7DW8-5 elicited dose-dependent local erythema at injection sites, but no evidence of systemic reactogenicity (fever, tachycardia, respiratory distress). 7DW8-5 provided a significant increase in IFN ELISPOT responses to both antigens, most significant at the 10g dose (fold increase in mean spot-forming units/million over no adjuvant: 9.8 and 5.8 for CSP and AMA1, respectively, p=0.04 for both). The magnitude of response did not correlate with circulating NKT cell level, indicating that this may be useful across broad populations. Initial decreases in circulating NKT cell levels on Day 1 normalized rapidly by Day 2. 7DW8-5 activated circulating plasmacytoid dendritic cells earlier than macaques who received vaccine alone, measured by CD40, CD80, and CD86 expression. Conclusion: 7DW8-5 provides a significant adjuvant effect on the cellular immunogenicity of an adenoviral vaccine in nonhuman primates, and is advancing into a Phase 1 clinical trial in healthy volunteers.

POSTERS 92 AIDS Vaccine 2011

Topic 02: Adjuvants P02.05


Co-Administration of NOD1 and NOD2 Ligands with HIV-1 p24 Increase Mucosal Immune Potency of Biodegradable Nanocarriers
V. Pavot1, N. Rochereau2, C. Genin2, E. Perouzel3, G. Tiraby3, T. Delaire4, S. Paul2, B. Verrier1
1 3

Posters
P02.06
Liposomes Containing Glucosyl Ceramide and the Adjuvant Monophosphoryl Lipid A Specifically Bind T4 Bacteriophage: A Self-Assembling Nanocarrier
K.K. Peachman1, O. Jobe1, L. Wieczorek1, L. Asher2, V.R. Polonis3, V. Rao4, C.R. Alving3, M. Rao3
1

IBCP-CNRS, Lyon, France; GIMAP, Saint Etienne, France; Invivogen, Toulouse, France; 4LMPB, Lyon, France
2

Background: The use of TLR ligands as mucosal adjuvant for vaccine administration is already largely described, but the use of NOD ligands is still investigated. As NOD ligands are able to induce production of pro-inflammatory proteins and chemokines, we have evaluated if their co-delivery with biodegradable nanocarriers carrying HIV Gag antigens amplify the mucosal immune responses in mice. Methods: We used Poly(Lactic Acid) (PLA) nanoparticles (200 nm) as vaccine vehicle for delivery of HIV-1 Gag p24. To assess their immunogenicity and the adjuvant effect of NOD agonists (NOD1 and NOD2), we have compared two routes of immunization, enteric and subcutaneous, in B6D2 mice with co-administration of NOD ligands. For each group, cellular and humoral responses have been analyzed on splenocytes and in vaginal washes, faeces and sera. Results: We first confirmed that PLA nanoparticles are efficient protein carriers of HIV p24 without alteration of the colloidal stability of the formulation. By analyzing humoral immune responses, we noticed that co-administration of p24-PLA NPs and NOD ligands through subcutaneous route was very efficient to induce higher anti-p24 IgG responses in rectal, vaginal washes and sera, irrespective of the sub classes. However, p24-specific IgA responses at the rectal site were only found after repeated enteric vaccination with adjuvanted formulations. Upon subcutaneous injection in mice, we could observe that p24PLA NPs co-administered with NOD1 ligand induced a significant decrease of the IgG1/IgG2a ratio compared to non-adjuvanted formulation meaning a Th1 orientation. Conclusion: When PLA are co-administered with NOD ligands as exemplified by the appearance of mucosal immunity, a coadjuvant effect was clearly observed. Use of NOD1 and 2 ligands as mucosal adjuvant deserve further experiments and we are currently investigating the mechanisms involved and increasing delivery efficacy after co-encapsulations.

U.S. Military HIV Research Program, Henry M. Jackson Foundation, Rockville, MD, USA; 2Walter Reed Army Institute of Research, Silver Spring, MD, USA; 3U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Rockville, MD, USA; 4The Catholic University of America, Washington, DC, USA
Background: Our goal was to create a self-assembling nanocarrier vaccine adjuvant formulation by utilizing bacteriophage T4 displaying HIV-1 gp41 antigens and liposomes containing glucosyl ceramide (GluCer) and monophosphoryl lipid A (MPLA). Liposomes containing MPLA have been demonstrated to have potent adjuvant activities for experimental vaccines both in humans and in animals and because of this the immunogenicity of the spontaneously bound nanocarrier antigen-adjuvant formulation was evaluated in small animals. Methods: A peptide antigen derived from either the C-trimer (Ct) or the c-terminal outer domain (COD) of HIV-1 gp41 was fused to the highly antigenic outer capsid protein (Hoc), a nonessential protein of T4 that spontaneously binds to the T4 capsid. This resulted in display of Ct-Hoc or COD-Hoc antigens on the T4 capsid. Mice and rabbits were immunized with Ct-Hoc-T4 and COD-Hoc-T4 constructs as such or bound to liposomes containing both GluCer and MPLA. The specificity of the spontaneously bound T4-liposomal formulation was determined by Biacore. Humoral responses in mice and rabbits and cellular immune responses in mice were analyzed. Results: The display of Ct-Hoc antigen on T4 capsids was confirmed by electron microscopy. Binding of phage T4 did not occur to glycolipids, such as galactosyl ceramide, containing an aldose in which the C-2 or C-4 conformations were not identical to glucose. These results strongly support previous reports that glucose is a major receptor moiety for T4 binding to Escherichia coli. High titer antigen-specific IgG antibodies were induced in the immunized mice and rabbits. Neutralizing antibodies were analyzed in the PBMC, TZM-Bl, and a macrophage neutralization assay. Cellular responses specific to both Ct and COD antigens were also induced in mice. Conclusion: Liposomes containing both GluCer and MPLA can spontaneously bind to T4 displaying HIV-1 gp41 antigens. This formulation could be utilized as an easily manufactured selfassembling antigen and adjuvant carrier.

AIDS Vaccine 2011

93

POSTERS

Posters
P02.07

Topic 02: Adjuvants P02.08


GM-CSF as an Adjuvant for MVA Vaccine: High Dose but Not Low Dose Inhibits T Cell Responses and Rectal IgA by Modulating DC Activation and Phenotype
R. Amara1, S. Kannanganat1, L. Lai1, S. Gangadhara1, S. Kwa1, P. Kozlowski2, V. Hirsch3, L.S. Wyatt3, P.L. Earl3, B. Moss3, H.L. Robinson4 Emory University, Atlanta, GA, USA; 2Louisiana State University Health Sciences Center, New Orleans, LA, USA; 3 National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA; 4Geovax Inc., Smyrna, GA, USA
1

Antigen-Specific Enhancement of Natural Human IgG Antibodies to Lipids Induced by a Liposomal Vaccine Containing Lipid A and a Protein Antigen
G.R. Matyas1, C.R. Alving1
1

Walter Reed Army Institute of Research, Rockville, MD, USA

Background: Monoclonal antibodies 2F5 and 4E10 not only bind to the protein epitope, but also to lipids. Multispecific antibodies with similar binding specificities to 2F5 and 4E10 and antibodies to lipids can readily be induced in mice by immunization with liposomes containing monophosphoryl lipid A (MPLA). However, it has never been demonstrated that antibodies with lipid binding specificity can be induced by vaccination of humans. This study was conducted to determine if antibodies to lipids can be induced or boosted in titer by vaccination of humans. Methods: ELISA for IgG to lipids was conducted on stored sera obtained from volunteers who received a candidate vaccine to Plasmodium falciparum (PNAS 1992; 89:358-362). The vaccine consisted of liposomes that contained both the recombinant protein antigen and MPLA as an adjuvant. Results: The pre-immune sera contained natural antibodies (NA) to one or more of the lipids in the vaccine: dimyristoyl phosphatidylcholine (DMPC), dimyristoyl phosphatidylglycerol (DMPG), cholesterol, and MPLA. Two weeks after the vaccine boost, increased levels of IgG to all of the liposomal lipids, especially DMPG and MPLA, were observed. Antibodies to phosphatidylinositol-4-phosphate (PIP) above the normal preimmune NA to PIP were also observed. Although PIP was not present in the immunizing liposomes, the anti-PIP antibodies were thought to represent a cross-reaction with DMPG. The immune response was antigen-specific in that NA to unrelated lipids that were not present in the liposomes, galactosyl ceramide and ganglioside GM1, were not increased by immunization. Furthermore, antibodies to beta-2-glycoprotein I or albumin were also no increased in titer following immunization. Conclusion: Antibodies to DMPC, DMPG, PIP, cholesterol, and MPLA can be induced or boosted in humans by immunization with liposomes containing MPLA. There were no adverse effects of these antibodies and no increases in the pathogenic antibodies to beta-2-glycoprotein I.

Background: We recently demonstrated that co-delivery of Granulocyte-Macrophage-Colony-Stimulating Factor (GM-CSF) DNA with SIV DNA vaccine enhances protection from acquisition of heterologous mucosal SIVsmE660 infection from 25% to 71% by a DNA/MVA vaccine. Here, we evaluate the optimal dose of GM-CSF for enhancing the immunogenicity and efficacy of MVA-only vaccine in rhesus macaques. Methods: A MMM regimen delivered MVA/SIV (108 plaque forming units, pfu) expressing SIV239 Gag, Pol and Env at weeks 0, 8 and 24. In addition, GM-CSF adjuvanted groups simultaneously received MVA expressing GM-CSF (GM) at indicated dose. There were 7-8 animals/group. Results: The MMM vaccine rapidly induced expression of CD80 (activation), CCR7 (lymph node homing) and alpha4 beta7 integrin (a4b7, gut homing) on plasmacytoid dendritic cells (PDC) and monocytoid DC (MDC) in blood. Consistent with DC activation, MMM vaccine elicited strong SIV-specific CD8 and CD4 T cell responses in blood, high titer and avidity Env-specific IgG in serum, and moderate levels of Env-specific IgA in rectal secretions. High doses (5x107 and 107 pfu) of GM inhibited SIV-specific T cell responses and SIV-specific IgA in rectal secretions whereas low doses (106 and 105 pfu) either enhanced or maintained them. However, high doses of GM did not inhibit SIV-specific IgG in serum. The high dose but not low dose GM groups inhibited CD80 expression on MDC and down regulated a4b7 on PDC that was strongly associated with inhibition of SIVspecific T cell responses and rectal IgA, respectively in the high dose GM groups. Conclusion: These results demonstrate that high doses of MVA/ GM-CSF inhibit cellular immune responses in blood and IgA in rectal secretions by modulating activation status of MDC and gut homing potential of PDC. These animals will be challenged with SIVsmE660 to study the influence of high and low dose GM-CSF on protection.

POSTERS 94 AIDS Vaccine 2011

Topic 02: Adjuvants P02.09


Polyethyleneimine Is a Potent Mucosal Adjuvant and Candidate Adjuvant for HIV-1 Vaccination
S. Brinckmann 1, F. Wegmann1, K. Gartlan1, Q. Sattentau1
1

Posters
P02.10
Degradable PEI Form as Safer Alternative for HIV1 Vaccine Development and Adjuvant Potency as a Common Trait of Oligoethyleneimines
S. Brinckmann 1, F. Wegmann1, K. Gartlan1, Q. Sattentau1
1

University of Oxford, Oxford, United Kingdom (Great Britain)

University of Oxford, Oxford, United Kingdom (Great Britain)

Background: There are no mucosal adjuvants licensed for use in man although protection against many mucosally-transmitted infections probably requires immune activation at the site of pathogen entry. Polyethyleneimine (PEI) is used extensively as a transfection reagent and in-vivo gene delivery vehicle. Here we show that PEI has potent mucosal adjuvant activity with HIV-1 glycoprotein. PEI recruits leukocytes, triggers a local balanced Th1/Th2 cytokine environment and targets antigen to lysosomelike compartments in antigen presenting cells. Methods: Balb/C mice were i.n. immunised with PEI (25 kD branched); PBS or CTB in formulation with HIV-1 glycoprotein (CN54gp140) antigen. Bleeds and vaginal lavages were obtained pre-prime, post-prime and post-boost immunisation and analysed for CN54gp140-specific IgG and IgA titres (ELISA). Spleens and cervical lymph nodes were obtained, restimulated with CN54gp140 and analysed for T cell proliferation (thymidine incorporation assay) and cytokine production (LUMINEX). Balb/C mice received i.p. injection of PEI, Alexa-647-ovalbumin, or both. Peritoneal lavages were obtained and cytokine levels determined (LUMINEX). Single cell suspensions were prepared and stained for leukocyte markers for flow cytometry. Naive peritoneal macrophages were incubated with Alexa488-CN54gp140 with or without PEI, fixed, permeabilised, stained with subcellular compartment markers for confocal microscopy. Results: PEI triggered local and systemic antibody levels of equal or higher size than gold standard adjuvant CTB. PEI also triggered T cell responses in spleen and lymph nodes of equal size and interestingly more localised character. Peritoneal administration showed that PEI recruits leukocytes, particularly monocytes, to the site of injection and triggers a local balanced Th1/Th2 cytokine environment. Analysis with Alexa 647- ovalbumin demonstrated that PEI targets antigen to antigen presenting cells. Confocal microscopy confirmed the presence of HIV-1 CN54gp140 inside macrophages when ex-vivo co-incubated with PEI. Conclusion: PEI has immunostimulatory properties and adjuvant potency for HIV-1 immunisation. PEI may merit development as a mucosal adjuvant for human use.

Background: There are no mucosal adjuvants licensed for use in man although protection against many mucosally-transmitted infections probably requires immune activation at the site of pathogen entry. Polyethyleneimine (PEI) is used extensively as a transfection reagent and in-vivo gene delivery vehicle. We have shown that PEI has potent mucosal adjuvant activity with HIV-1 glycoprotein. One of the potential drawbacks of PEI is its potential toxicity. In these studies, we compared various PEI forms including degradable PEI forms for their adjuvant potency and toxicity. Methods: Various linear and branched, degradable and non-degradable PEI forms were used in formulation with HIV-1 glyprotein (CN54gp140) antigen for i.n, i.p.. and s.c. immunisation in female Balb/c mice. Bleeds and vaginal lavages were obtained at regular intervals pre-prime, post-prime and post-boost immunisation. Body weights were recorded at 0, 6, 12, 24, and 48 hours post immunisation. Lavage and serum samples were analysed for CN54gp140 specific IgG and IgA titres using ELISA. Results: A wide range of structural PEI forms showed enhanced local and systemic antibody responses when co-formulated with CN54gp140 for immunisation in mice via various administration routes. Interestingly, no relation was found between molecular weight (mW) or linear/branched structure and adjuvant potency, which is in contrast to PEI gene transfection studies that showed optimal transfection for high mW branched forms. Some degradable forms in our studies showed substantial adjuvant potency combined with lower observed body weight loss in mice. Conclusion: PEIs immunostimulatory properties are a shared trait between various structural, degradable and non-degrable PEI forms. Some degradable PEI forms triggered substantially less weight loss in mice, indicative of a lower toxicity profile, while still being potent adjuvants in these models. Degradable PEI forms could therefore form a safer alternative for standard non-degradable forms when developing PEI as an adjuvant for HIV-1 vaccination.

AIDS Vaccine 2011

95

POSTERS

Posters
P03.01

Topic 03: Animal Models P03.02 LB


HIV Envelop Proteins Immunization Effect in Hu-SCID Mice Model
A. Bychenko1, G. Kuznecovs2, N. Polyakov3, A. Galkin4, E. Gagarina4, E. Filinova5
1

Differential Expression of Transcription Factors in SIV-Specific CD8+ T Cells at Week 5 and Week 20 After Vaccination with SIV239nef
J.M. Billingsley , P.A. Rajakumar , N.C. Salisch , Y.V. Kuzmichev , M.A. Connole1, R.K. Reeves1, R.P. Johnson1
1 1 1 1

New England Primate Research Center Harvard Medical School, Southborough, MA, USA
1

Background: Current evidence suggests that protective immunity against vaginal challenge in SIVnef-vaccinated macaques develops at 20 weeks after vaccination, whereas the magnitude of SIV-specific CD8+ T cells peaks at 5 weeks. Phenotypic analyses indicate a maturation of memory responses from week 5 to 20, as characterized by upregulation of CCR7 and CD127. These observations suggest that the quality of a more mature CD8+ T cell response, as opposed to the magnitude, may correlate with protection. We therefore analyzed differential expression of transcription factors involved in T cell maturation in SIV-specific CD8+ T cells at 5 and 20 weeks after SIVnef vaccination. Methods: Highly parallel qRT-PCR (Fluidigm) was used to characterize the expression of 23 transcription factors in CD8+ T cells sorted into nave, central, transitional, and effector memory subsets, as well as in SIV-specific CD8+ T cells obtained at wk5 and wk20 after SIV239nef vaccination. Results: Unsupervised hierarchical clustering segregated nave and memory cell populations by phenotypic class. While the expression of some transcription factors, e.g. T-bet and Blimp-1, progressively increased during memory cell differentiation, others, (TCF7 and LEF1), progressively decreased. Further, SIVspecific CD8+ cells segregated into wk5 and wk20 clusters, with the wk 20 population exhibiting increased levels of transcription factors associated with both quiescent memory T cells (TCF7, BAZF) and maintenance of effector function (Eomes, T-Bet). Conclusion: Our data indicate distinct transcriptional profiles of the different memory CD8+ T cell subsets, as well as clear qualitative differences in wk5 and wk20 SIV-specific CD8+ T cell transcriptomes. We further demonstrate that the mature CD8+ T cell response induced by SIV239nef is characterized by the expression of transcription factors associated with both central memory and effector memory T cells. Analysis of transcription factor expression therefore provides a valuable complement to the analysis of memory cell differentiation based on classical phenotypic markers.

Advanced Biomedical Research Laboratory, Moscow, Russian Federation; 2Advanced Biomedical Research Laboratory, Physics Method of Analysis, Moscow, Russian Federation; 3 Bioorganic Chemistry Institute Named by Shemyakin and Ovchinnikov, Moscow, Russian Federation; 4Advanced Biomedical Research Laboratory, Molecular Biology/Genetics De, Moscow, Russian Federation; 5Advanced Biomedical Research Laboratory, Cellular Biology/Virology Department, Moscow, Russian Federation
Background: Immunizations and challenging of the immune deficient SCID mice capable for engraftment with human immunocompetent cells (Hu-SCID-mouse) as an animal model for the evaluation of the effectiveness of an HIV vaccine are described Methods: Envelop proteins gp120-gp160 were isolated from 1.5-2 litres HIV-1 subtype A U455 and PokA-79 strains MT4- or PBMC-6-weeks in vitro cultivation (playbacks) culture and identified with MS-MS. Immunogenic compositions were tailored and isolated in/from L. tarentolae expression system. HuSCID mice engrafted with single donor PBMC (peripheral blood mononuclear cells) and with DC (dendrite cells) immunocompetent cells were immunized sub-cutaneously with recombinant gp120gp160 cocktails and challenged intraperitoneally with U455 and PokA-79 strains and their playbacks. HIV laboratory strains and PBMC-DC- playbacks challenging viral load as well as vaccination efficiency were measured in blood serum by RealTime PCR. HIV-specific immune response in blood serum was carried out by p120-p160 Results: U455 and PokA-79 HIV-1 laboratory strains multiplied successfully in Hu-SCID mice engrafted with lymphocyte MT-4 or monocyte U937 cell culture backgrounds with titres 104-108 copies/ml but were unable to provide bloodstream viral load in immune-competent Hu-SCID mice engrafted with PBMC or DC background. In vitro controls showed HIV RNA 1010-1013 c/ml for U937-MT-4-cultivated HIV laboratory strains but negative dynamics with RNA 104-105 c/ml for DC- or first passages PBMCcultivated strains. TCID50 data for in vitro controls followed the same ratio. However after 5-6 weeks of PBMC-background in vitro cultivation playbacks RNA c/ml titre and TCID50 data rise, show positive multiplication dynamics and gp120 sequence variability widening. Playback strains demonstrated positive bloodstream titres in Hu-SCID-PBMC mice. Recombinant gp120-160 cocktails tailored on base of playbacks envelop mapping used for HuSCID-PBMC immunizations blocked HIV challenging in these Conclusion: In vitro controls suggest HIV gp120-gp160 infection-responsible variability is determined by background human cells receptors diversity crucial for virus recognition, invasion and challenging

POSTERS 96 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.01


Putative Rhesus Macaque Germline Antibodies of Broadly HIV-Neutralizing Antibodies: Similarities and Differences Compared with the Human Counterparts
M. Zhang , T. Yuan , J. Li , Y. Zhang , Y. Wang , E. Streaker , D.S. Dimitrov2
1 1 1 1 2 3 1

Posters

P04.02
Genetic and Neutralization Sensitivity of Diverse HIV-1 Env Clones from Chronically Infected Patients in China
H. Shang1, X. Han1, X. Shi2, X. Wang2, L. Zhang2 Key Laboratory of AIDS Immunology of Ministry of Health, Shenyang, China; 2AIDS Research Center, School of Medicine, Tsinghua University, Beijing, China
1

The University of Hong Kong, Pokfulam, China; 2National Cancer Institute at Frederick, NIH, Frederick, MD, USA; 3 National Cancer Institute-Frederick, National Institutes of Health, Frederick, MD, USA
Background: Broadly neutralizing antibodies (bnAbs) are likely to be a key component of protective immunity conferred by an effective HIV-1 vaccine. We and others have reported that putative human germline predecessors of bnAbs lack measurable binding to the HIV-1 envelope glycoprotein (Env) that could be a new challenge to elicit human bnAbs. Rhesus macaques have been used as a nonhuman primate model for testing HIV-1 vaccine candidates, but little is known about their germline antibodies. Methods: We used one of the several best characterized bnAbs, b12, as a model Ab and identified a putative rhesus macaque b12 germline predecessor by sequence analysis. We compared the human and macaque germline predecessors and possible intermediate antibodies of b12 by sequence analysis and by ELISA and neutralization assays. Results: Putative rhesus macaque b12 germline predecessor also did not show measurable binding to HIV-1 Envs. However, we found different sequence characteristics between the human and macaque germline genes and their intermediate antibodies of b12 isolated from B cell receptor repertoires of nonimmune rhesus macaques and human. Conclusion: These results suggest that initiation of somatic maturation of germline b12 predecssor in rhesus macaque may also be a challenge. But maturation pathways leading to elicitation of b12 or b12-like antibodies in rhesus macaques may be different from those in human, suggesting that primary immunogens to initiate somatic maturation and Env-based vaccine immunogens to further elicit bnAbs in rhesus macaque may be different from those in human. This finding may have implications for HIV-1 vaccine development.

Background: As HIV-1 continues to spread in China from traditional high-risk populations to the general public, its genetic makeup has become increasingly complex. However, the impact of these genetic changes on the biological and neutralization sensitivity of the virus is unknown. The current study aims to characterize the genetic, biological and neutralization sensitivity of HIV-1 identified in China between 2004 and 2007. Methods: Full-length envelope genes were amplified by PCR directly from proviral DNA extracted from patients unclutured PBMC and cloned into expression vector. Env-bearing pseudotyped viruses were generated by co-transfection of env-expressing plasmid together with backbone construct pNL43R-E-luciferase into the 293 cells. Neutralization sensitivity of these diverse HIV-1 to subtype-specific plasma pools from infected patients as well as to broadly neutralizing monoclonal antibodies (2F5, 4E10, 2G12, PG9, PG16, IgG1b12, and VRC01 ) were analyzed. Results: Pseudotyped viruses built upon the viable env genes have demonstrated their substantial variability in mediating viral entry, and in sensitivity to neutralization by subtype-specific plasma pools and broadly neutralizing monoclonal antibodies (bnmAb). Many viruses are resistant to one or more bnmAb including those known to have high potency against diverse viruses from outside China. Sequence and structural analysis has revealed several mechanisms by which these resistant viruses escape recognition from bnmAb. Conclusion: We believe that these results will help us to better understand the impact of genetic diversity on the neutralizing sensitivity of the viruses, and to facilitate design of immunogens capable of eliciting antibodies with similar potency and breadth as bnmAb. The viruses characterized here will also provide a strong foundation for establishing a broader and more representative Chinese virus panel to evaluate the antibody responses in infected and vaccinated individuals, and to contribute to the international virus panel where only a limited number of Chinese viruses are currently available

AIDS Vaccine 2011

97

POSTERS

Posters
P04.03

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.04


In Vitro Neutralization Kinetics of the Human Monoclonal Antibody IgG1 b12 Which Is Protective In The SHIV Rhesus Macaque Challenge Model.
D. Davis2, W. Koornstra2, J. Heeney1, W. Bogers2
1 Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom (Great Britain); 2 Biomedical Primate Research Centre, Rijswijk, Netherlands

Superinfection Results in Potent Neutralizing Antibody Responses to the Superinfecting Virus, but Does Not Necessarily Enhance Neutralization Breadth
D.J. Sheward1, P.L. Moore2, M.C. Madiga2, E.S. Gray2, R. Ntale1, Z.L. Woodman3, J. Bhiman2, H. Harvey1, S. Sibeko4, K. Mlisana4, S.S. Abdool Karim4, L. Morris2, C. Williamson1
1

University of Cape Town, Cape Town, South Africa; 2AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa; 3Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa; 4Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa

Background: A better understanding of how neutralizing antibody responses develop during natural infection and the factors that augment them would be invaluable in the design of improved immunogens and immunization protocols. HIV dual infection (infection by >1 distinct HIV strain) provides a unique opportunity to evaluate whether polyvalent immunogens and prime-boost strategies are likely to enhance the breadth and potency of vaccine-induced responses against HIV. Methods: Ten dual-infected participants were identified from the CAPRISA002 cohort, 5 of whom were co-infected at or prior to seroconversion, and 5 were superinfected within the first year of infection. Neutralization breadth at 3 years postinfection was compared to 16 singly-infected participants using a panel of 44 heterologous viruses, including subtypes A, B, and C. Autologous plasma neutralizing titers in 3 superinfected, and 3 co-infected individuals were assessed longitudinally using representative clones from multiple timepoints in the pseudovirus-based TZM-bl assay. Results: There was no association between co-infection, superinfection, or diversity in early infection and the development of greater neutralization breadth. Furthermore, titers to the primaryinfecting variants were not boosted following superinfection. However, 2 of the 3 superinfected participants developed elevated titers against the superinfecting virus, with ID50 values exceeding 1:20,000 and 1:40,000 respectively. In one individual these high titers were generated despite maintaining low viral loads, frequently below detection (<400 copies/ml). None of the 3 coinfected participants generated similarly elevated titers, suggesting that sequential infection was a driving factor. Conclusion: The lack of an association between dual-infection and breadth highlights the fact that polyvalent immunogens may not necessarily improve the breadth of elicited humoral responses. However, the high titers generated against the superinfecting variants suggest that a prime-boost approach has the potential to increase the potency of neutralizing responses against the boosting immunogen. As existing specificities were not boosted, the mechanism is likely distinct from the anamnestic response.

Background: In passive transfer experiments, the human monoclonal antibody IgG1 b12 protects rhesus macaques against infection with both the neutralization sensitive SHIVSF162P4 and the more resistant SHIVSF162P3. Relatively high concentrations of antibody are required to protect macaques against a bolus while lower quantitites are protective in the repeated, low dose, challenge model. Methods: HIV-1 SF162, SHIVSF162P4 and SHIVSF162P3 were grown in human PBMCs. Virus was mixed with antibody and incubated. Aliquots of the mixture were exposed to GHOST cells. Cultures were washed to terminate the absortion phase. Infected cells were quantified by FACS. Results are expressed either as plots of reductions in infectious virus over time or residually infectious virus against virus dose. Results: Neutralization of both HIV-1 and SHIV is exponential during both the incubation and absorption phases. However, at relatively high doses of antibody, there is significant neutralization without prior incubation. Dose response plots of infected cells against virus dose are linear with a gradient of one passing through the origin. Incubation of low doses of virus with a low concentration of antibody yields a dose response plot which is parallel to controls: the percentage neutralization is low with 100 TCID50 virus but increases as the virus dose is reduced, reaching complete loss of infectivity at the point where the plot crosses the horizontal axis. Conclusion: IgG1 b12 forms a complex with virus. However, this complex is still infectious. Events during the absorption phase of a SHIV neutralization assay determine the number (rather than the proportion) of viruses which subsequently lose infectivity. At low concentrations of antibody a limited number of infectious virus can be completely inactivated. It is proposed that this number is proportional to the dose of virus which a vaccinated individual can be exposed to without subsequent infection. Such assays should be used to monitor human vaccine trials.

POSTERS 98 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.05


Broad ADCC Responses and ADCC Directed at VPU Epitopes in Humans Is Associated with Slow Progression of HIV
L.H. Wren1, M. Navis1, A. Chung1, G. Isitman1, A.D. Kelleher2, I. Stratov1, S.J. Kent1
2

Posters

P04.06
Activity of Broadly Neutralizing Antibodies Against Recently Transmitted Subtype B HIV-1 Variants from Early and Late in the Endemic
Z. Euler1, E.M. Bunnik1, J.A. Burger1, B.D. Boeser-Nunnink1, M.L. Grijsen1, J.M. Prins1, H. Schuitemaker1
1

The University of Melbourne, Melbourne, Australia; National Centre for HIV Epidemiology and Clinical Research, Sydney, Australia
1

Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands

Background: Robust antibody-dependant cellular cytotoxicity (ADCC) responses at the site of HIV infection may enable the elimination of virus prior to viral spread and latency. Identifying the targets of ADCC responses in long-term slow progressors (LTSP) could help direct the future design of vaccines to elicit beneficial ADCC responses. Methods: A cohort of 61 LTSPs and a cohort of 78 unselected subjects with varying rates of HIV progression were analysed for ADCC responses against gp140 proteins using cytolysis-based and NK-cell activation-based assays. ADCC Responses against HIV-1 peptide pools were also analysed and specific epitopes mapped using an intracellular cytokine staining assay to detect NK cell activation. Results: HIV-specific ADCC responses correlated with reduced loss of CD4 T cells across the two cohorts (p=0.027). Broader ADCC responses against a greater number of HIV-1 peptide pools were found in subjects of the LTSP cohort than in the unselected cohort (p=0.006). In addition, ADCC responses against epitopes within the Vpu protein were overrepresented in slow progressors and two key ADCC epitopes in VPU were observed. Conclusion: Broad ADCC responses correlate with slower progression of HIV. We hypothesise that inducing ADCC Abs against specific epitopes within multiple HIV-1 proteins, including regulatory proteins such as VPU may provide a mechanism for protection from acquisition of HIV-1 or slower disease progression of those that do acquire HIV-1. Further work to design vaccines that elicit ADCC against key targets needs to be urgently explored.

Background: For the development of a neutralizing antibodybased HIV-1 vaccine, it is important to characterize which antibody specificities are most effective against currently circulating HIV-1 variants. As we recently reported that HIV1 has become more resistant to antibody neutralization over the course of the epidemic, we here explored if this increased neutralization resistance was also observed for the newly identified broadly neutralizing antibodies (BrNAbs) PG9, PG16, and VRC01. Furthermore, we performed a comprehensive analysis of the neutralizing sensitivity of currently circulating recently transmitted subtype B viruses to the currently most known BrNAbs. Methods: We determined the neutralizing sensitivity to PG9, PG16, and VRC01 of virus variants that were isolated less than six months after seroconversion from individuals in the Amsterdam cohort who seroconverted between 1985 and 1989 (n = 14), as well as b12, 2F5,4E10, 2G12 for individuals who seroconverted between 2003 and 2006 (n = 21). Moreover, we looked at potential mutations that would influence neutralizing sensitivity. Results: Viruses from contemporary seroconverters were significantly more resistant to neutralization by VRC01 and tended to be more resistant to neutralization by PG16 than historic seroconverters, which coincided with the presence of more mutations at positions in the viral envelope that may potentially influence neutralization by these antibodies. Despite this increased neutralization resistance, all recently transmitted viruses from contemporary seroconverters were sensitive to at least one BrNAb at concentrations of 5 g/ml, with PG9, PG16, and VRC01 showing the greatest breadth at lower concentrations. Conclusion: Our results show that while HIV-1 has become more resistant to CD4-binding site-directed neutralization over the course of the epidemic, all virus variants were sensitive to at least one of the BrNAbs tested. These results suggest that a vaccine capable of eliciting multiple BRNAb specificities will be necessary for protection of the population against HIV-1 infection.

AIDS Vaccine 2011

99

POSTERS

Posters
P04.07

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.08


Broad ADCC Responses and ADCC Directed at VPU Epitopes in Humans Is Associated with Slow Progression of HIV
D.V. Almeida 1, V. Bongertz1, F.H. Crtes1, M.L. Guimares1, J.H. Pilloto1, G. Bello1, V. Veloso2, M.G. Morgado1
1

Neutralizing Activity Against Both gp120 and gp41 in Patients with HIV-Specific Cross-Reactive Neutralizing Humoral Immunity
M.J. van Gils1, E.T. Crooks2, J.M. Decker3, E.S. Gray4, N.L. Tumba4, R. Lynch5, J.R. Mascola5, L. Morris4, J. Binley2, H. Schuitemaker1 Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands; 2Torrey Pines Institute for Molecular studies, San Diego, CA, USA; 3Department of Medicine, University of Alabama, Birmingham, AL, USA; 4National Institute for Communicable Diseases, Johannesburg, South Africa; 5Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
1

Fundation Osvaldo Cruz, Rio de Janeiro, Brazil; 2Evandro Chagas Clinical Research Institute, Rio de Janeiro, Brazil

Background: To date, no HIV-1 immunogen is available that can elicit a potent cross-reactive neutralizing humoral immune response. One of the current approaches is the identification, characterization and subsequent use of epitopes of very potent broadly neutralizing antibodies as immunogens to elicit HIV-1 specific neutralizing antibodies with similar potency and breadth. Methods: In an attempt to identify novel epitopes for broadly neutralizing antibodies, we selected five individuals who at two years post-imputed-SC had developed potent serum crossreactive neutralizing activity (CRNA). Results: Interestingly, sera from all five patients to some extent had post-CD4-CCR5-binding HIV-1 neutralizing activity (IC50 titers range 1:100-1:350; 20-70% of neutralization activity), implicating the presence of anti-MPER antibodies. Chimeric HIV-2 variants with HIV-1 MPER or parts thereof, were used to identify the exact epitopes of the anti-MPER activity, with all five patients targeting different parts of the MPER-region. Adsorption of MPER antibodies with linear-MPER-coated beads reduced CRNA in sera from two individuals, however the contribution of linear anti-MPER activity to the CRNA in the other three patients could not be confirmed. In all five sera, detectable anti-gp120 activity was measured, although it did not reach more than 50% of the neutralization capacity. Assays so far indicate little if any evidence for neutralizing antibodies similar to b12, 2G12, PG9/ PG16, and CD4i or anti-core antibodies, although there was evidence for what may be type-specific N332A-sensitive activity. Neutralizing antibodies to the CD4-binding site are currently under investigation in these patients. Conclusion: These results show that our sera all contain significant titers of anti-gp41 MPER neutralizing antibodies. However, there is evidence for other epitopes to be targeted by the CRNA in these patients, including those directed to gp120, suggesting that cross-reactive neutralization is achieved by multiple specificities. Further characterization of the crossreactive neutralizing epitopes may assist in the development of new HIV-1 vaccine targets.

Background: Tests for the detection of humoral immune response to HIV-1 have to be established, demanding regional efforts to permit assessments of efficacy of future vaccine trials and allow comparisons on a global level. Our goal is to use the TZM-bl assay to investigate Neutralization antibody (NAb)able to inhibit HIV-1 replication in plasma from HIV-1 positive individuals with distinct profiles of Aids progression. Methods: Eight env-pseudoviruses were evaluated against 22 plasmas from HIV-1-infected individuals: 13 typical-progressors (TP) and 9 long-term non-progressors (LTNP). We used as a control sCD4 and 2F5 Mab and 3 clade/variant plasma pooled TP (B,B/Bbr,F). The same plasmas were tested against the HIV-1 IIIB isolate in primary lymphocyte (ID90%). Results: From the 13 TP plasma tested, 2 (15%) were potent and broad in their neutralizing capacity (100%), as well as the HIV-1 IIIB isolate (PBMC), 7 (53%) neutralized 50% of the env-pseudotyped tested, the remain varied from 25-30%. In the ID50% in TZMBL assay 104 titer points (13 TP plasmas x 8 pseudoviruses)were evaluated, from those 64 (62%) had neutralizing titers from 1:20-1:4374 and 40 (38%) havent NAb. Of the 72 (9 plasmas x 8 pseudoviruses) points LTNP plasma tested, 10 (14%) had neutralizing titers between 1:50-1:6250 and 62 (86%) havent NAb. The highest neutralization titer was verified in the B/Bbr1 pooled. Conclusion: Neutralization assay in TZM-bl cells was successfully reproduced in the laboratory being a tool for the detection of NAb from TP and LTNP plasmas in distinct proportions, showing a high capacity to neutralize virus/pseudovirus. These results and these reference reagents enable the participation of Brazil in future assays of neutralizing antibodies induced by vaccine to HIV-1. Financial support: GHVE and Dpto DST/AIDS and Hepatites MS.

POSTERS 100 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.09


Development of Broadly Neutralizing Antibody Responses in a Large Sub-Sahara African HIV Primary Infection Cohort
E. Landais1, M.D. Simek2, S. Lakhi3, E. Karia3, A. Kamali4, M. Inambao3, E.J. Sanders5, T. Wrin6, E. Cormier7, M.A. Price8, P.N. Amornkul8, P. Poignard1, The IAVI African HIV Research Network1 International AIDS Vaccine Initiative Neutralizing Antibody Center, La Jolla, CA, USA; 2International AIDS Vaccine Intitiative, New York, NY, USA; 3Rwanda Zambia HIV Research Group, Lusaka/Ndola/Kigali, Zambia; 4MRC/UVRI Uganda Research Unit on AIDS, Masaka and Entebbe, Uganda; 5 Centre for Geographic Medicine-Coast, Kenya Medical Research Institute, Kilifi, Kenya; 6Monogram Biosciences, Inc, South San Francisco, CA, USA; 7International AIDS Vaccine Initiative Human immunology laboratory, London, United Kingdom (Great Britain); 8International AIDS Vaccine Initiative, San Francisco, CA, USA
1

Posters

P04.10
FACS-Based Epitope Mapping Platform to Characterise New Identified Monoclonal AntiHIV-1 Envelope Antibodies
A. Kliche1, R. Wagner1
1

University of Regensburg, Regensburg, Germany

Background: Recent systematic approaches for identification of HIV-1 infected patients with neutralizing sera and subsequent generation of new broadly neutralizing monoclonal antibodies (bNMAb) create the need for a rapid characterisation tool which allows the determination of conformational epitopes on a native envelope structure. Methods: A sequentional permutation envelope library based on the isolate ZM96 was generated, where every amino acid is stepwise mutated to an alanine, resulting in 608 variants with a single point mutation. Every variant is used in a separate transient transfection and analysed by high throughput FACS analysis using an antibody of interest in combination with a second gp41 antibody used as an internal standard. First the screening process was validated using the v3 antibody HGN194 known to recognize a linear epitope as well as soluble CD4 which binds to an highly conformational epitope. Second, new antibodies with so far unmapped epitopes like HJ16 were screened. Results: The screen with HGN194 showed several critical amino acids for binding in the v3 region and confirms the published data by Corti D. at al. 2010. The screen using sCD4 confirmed several known amino acid positions which are known to be critical for the binding of the receptor to the envelope. The screening of HJ16 resulted in the identification of the several critical amino acids within the gp120 envelope for the binding of the bNMAB. These positions suggest a high conformational epitope. Additional mutations were identified which showed an enhanced binding of the bNMAB. Confirmed amino acid positions were modelled in the gp120 core structure. Conclusion: This method allows the identification of conformational epitopes of anti-HIV envelope antibodies on a native, trim-eric and membrane bound envelope.

Background: Recent studies have demonstrated that 10% to 20% of individuals infected with HIV-1 are capable of generating HIV-1 broadly neutralizing antibodies (bNAbs). Understanding the correlates of how these broad responses develop could be extremely important for the design of a protective vaccine. Methods: The IAVI Protocol C program investigates a longitudinal cohort of primary HIV-1 infection in Eastern and South Africa. Currently over 400 donors with recent HIV-1 infection have been recruited. To date, development of neutralization has been evaluated on 200 individuals, including 174 with at least 3 years of follow-up. Neutralization activity was assessed at Monogram Biosciences using a 6 cross-clade pseudo-virus panel highly predictive of neutralization breadth on larger panels. Neutralization of an isolate was recorded positive if equal or greater than 40% at a 1/100 serum dilution. Results: At year 2, only 4 individuals out of 171 had developed broad neutralizing activity, with 5 out of 6 viruses neutralized. In contrast, at year 3, 16 donors out of 163 (9.8%) neutralized more than 4 viruses. In addition, 69 individuals (36%) neutralized 3 or 4 viruses on the panel. Broad responses developed at year 4 in 9 additional donors out of 71 (12.7%). In most individuals, neutralization breadth developed simultaneously, appeared to be relatively stable with time, and no significant improvement of potency was noted over time. Screening of an additional 96 donors is pending. Conclusion: In conclusion, our current data show that in our large longitudinal sub-Sahara African cohort, broad neutralizing responses have so far developed in about 15% of individuals and essentially at year 3. Analysis of correlates of breadth development with clinical parameters, such as CD4 counts and viral load, is ongoing as well as the mapping of broad neutralizing specificities in the top neutralizers.

AIDS Vaccine 2011

101

POSTERS

Posters
P04.11

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.12


Single Mutation Turns a Non-Binding Germlinelike Predecessor of Broadly Neutralizing Antibody into a Binding Antibody to HIV-1 Envelope Glycoproteins
T. Yuan1, M. Zhang1
1

Innate Immune Signals Regulate Non-Neutralizing Effector Functions of Antibodies Through Glycosylation
A. Mahan1, J. Robinson2, G. Alter3 Harvard University, Charlestown, MA, USA; 2Tulane University, New Orleans, LA, USA; 3Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA
1

AIDS Institute, The University of Hong Kong, Hong Kong, China

Background: Data from the RV144 trial have shown that the modest success of this trial was not dependent on elicitation of CD8+ T cell or neutralizing antibody responses. Interestingly, most vaccinees induced non-neutralizing antibodies, which raises the possibility that other humoral antiviral activities may have contributed to protection. In fact, innate immune recruiting antibodies are enriched in long-term non-progressors, and this humoral activity has been shown to play a critical role in promoting the sterilizing protective effect of some neutralizing antibodies. However, the mechanisms by which these effector antibodies can be induced in vivo through vaccination, are not understood. We hypothesized that innate immune signals, particularly toll-like receptor (TLR) ligands, are able to modulate effector functionality of antibodies. Methods: Gene expression of a set of over 20 glycosylation enzymes was analyzed in healthy bulk B cells stimulated with TLR ligands to identify interesting changes in glycan structure. Results: Specific TLR stimulation of bulk B cells resulted in significant alterations of glycosylation enzyme expression. These changes related to the subcellular localization of the TLRs: extracellular TLRs did not induce significant changes in glycosylation enzyme expression, however, intracellular nucleic acid sensing TLRs 3, 7, 8, and 9 caused significant alterations. Interestingly, stimulation of the nucleic acid sensing receptors that recognize intracellular pathogens, such as HIV, significantly altered glycosylation gene expression. These changes correlated with a decrease in galactose, fucose, and sialic acid additions, which may correspond to the induction of more inflammatory, less branched glycan structures, that mediate stronger effector functions. Conclusion: These data are the first to show that innate immune signals modulate the antibody glycan patterns on stimulated B cells toward more inflammatory structures. This study strongly suggests that specific signals may be exploited in future vaccines to elicit antibody responses with specific glycosylation patterns and tailored effector functions.

Background: Broadly neutralizing antibodies (BnAbs) against human immunodeficiency virus type I (HIV-1) are rare in natural infection. We and other groups have reported that HIV-1 bnAbs are highly diversified from their germline-like predecessors. We do not know what are the minimum mutations required for converting non-binding germline-like predecessors to Envbinding antibodies. Methods: We started with the bnAb b12 as a prototype and generated six chimeric scFv b12 variants by sequentially replacing the heavy chain V-segment (HV), D(J)-segment [HD(J)], and the light chain variable region (VL) in b12 germline-like predecessor with the mature counterparts, and tested the scFv variants for binding and neutralizing activities in ELISA and pseudovirus assays. Results: A point mutation in germline heavy chain D-segment from Y to D converted nonbinding germline-like b12 to an Env-binding antibody. Replacement with either mature HV or mature VL also made the germline-like b12 bind to Env, but none of the single segment replacements conferred neutralization ability to the germline antibody. Mature VL in combination with mature HD(J), or mature HV, or both conferred increasing neutralization activity to the germline antibody. Hybrid scFv, mature VH / germline VL, did not neutralize the virus. Conclusion: During antibody maturation, less mutation, as low as one single nucleotide change, may be needed to turn a non-binding germline-like predecessor of bnAbs into a binding antibody intermediate to HIV-1 Env, which may further mature to bnAbs upon HIV-1 infection or vaccination. Somatic maturation in HV-, HD(J)-segments and VL of germline-like b12 additively contributed to the binding of b12 to Envs. Although b12 light chain did not make contact with gp120 core in the co-crystal structure, mature b12 VL was required for chimeric b12 variants to neutralize the virus. These results may have implications for vaccine development.

POSTERS 102 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.13


B Cell Depletion in HIV-1 Subtype A Infected Ugandan Adults: Relationship to CD4 Count, Viral Load and Humoral Immune Responses
P.O. Oballah1, B. Flach1, L.A. Eller2, M.A. Eller2, B.J. Ouma1, M.d. Souza3, D. Guwatudde1, F. Wabwire-Mangen4, B.K. Brown2, N.L. Michael2, M.L. Robb2, D. Montefiori5, V.R. Polonis2
1

Posters

P04.14
Llama Antibody Fragments that Potently Neutralize HIV-1 at the CD4 Binding Site of HIV-1 gp120
L. McCoy1, A. Forsman1, B. Bulmer-Thomas1, N. Strokappe2, T. Verrips2, L. Chen3, P.D. Kwong3, R.A. Weiss1
1

Makerere University Walter Reed Project, Kampala, Uganda; 2 U.S. Military HIV Research Program, Rockville, MD, USA; 3 Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 4Makerere University School of Public Health, Kampala, Uganda; 5Duke University, Durham, NC, USA
Background: HIV-1 infection leads to B cell dysfunction through mechanisms that are not fully characterized, particularly in subjects infected with non-B subtypes. To better understand the nature of B cell dysfunctions in East African subtype A infection, the relationship between B cell depletion, antibody function and disease progression was assessed. Methods: Participants were recruited under an IRB approved vaccine cohort protocol. Enumeration of lymphocyte subsets was performed using FACS MultiTEST, TruCount tubes and a FACSCalibur flow cytometer. Plasma viral load was measured by the Roche HIV-1 Monitor Test version 1.5. Sera from 50 subjects with chronic subtype A infection were tested by ELISA for gp120 binding antibodies and in the TZM-bl neutralization assay against a panel of 10 pseudoviruses from newly transmitted infections of subtypes A-D and CRF02_AG. Results: B cell numbers were significantly lower in HIV-1+ patients, compared to community matched HIV-negative controls (p<0.0001). Neutralization and binding antibody titers showed no correlation with viral load or CD4 counts. However, B cell absolute counts were found to correlate inversely with neutralizing titers against subtype A and CRF02_AG viruses. A positive correlation was observed between subtype A gp120 binding titers and neutralizing antibody breadth (p < 0.02) and titer (p < 0.05). Additionally, subtype A sera showed preferential neutralization of subtype A and CRF02_AG pseudoviruses, compared with pseudoviruses expressing non-A envelopes (p<0.0001). Conclusion: In patients with chronic HIV-1 subtype A infection, significant B cell depletion can be observed which does not appear to be associated with a decrease in functional antibodies. The inverse correlation between functional antibody titers and B cell numbers may be due to B cell maturation and migration out of the periphery. These findings also further substantiate the importance of subtype in the specificity of cross-clade neutralizing antibody responses in HIV infection.

University College London, London, United Kingdom (Great Britain); 2University of Utrecht, Utrecht, Netherlands; 3 Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
Background: Llamas produce heavy-chain only antibodies, from which the small (~15kd) binding fragment (VHH) may readily be cloned into a phagemid vector to construct phage libraries. We have isolated a number of VHH from libraries derived from llamas immunized with recombinant gp120 or trimeric gp140 which exhibit strong neutralization of diverse strains of HIV-1. Methods: Individual VHH clones expressed in E.coli from a phagemid vector were screened for binding to recombinant HIV1 envelope proteins and neutralisation ability against HIV-1 in the TZM-bl cell-based assay. Results: The best VHH isolated to date, J3, neutralizes almost 100% of HIV-1 pseudoviruses tested, with a breadth encompassing clades A, B, C, A/G and B/C. Structural studies of VHH:gp120 complexes indicate that both the angle at which VHH bind to their target and their small size compared to entire antibodies contribute to their neutralizing properties. Conclusion: Our studies of llama VHH are useful in three aspects of protection against HIV infection: (a) they show that experimental immunization with recombinant HIV envelope can elicit broad neutralizing responses; (b) they indicate that neutralization epitopes can be exploited in immunogen design for B-cell based vaccines; (c) the VHH themselves can be developed as potential microbicides owing to their stability over a wide range of temperature and pH, low immunogenicity and ease of scale-up for inexpensive mass production.

AIDS Vaccine 2011

103

POSTERS

Posters
P04.15

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.16


HIV-1 Envelope Characteristics that Coincide with the Development of Cross-Reactive Neutralizing Activity in HIV-1 Infected Patients
T.L. van den Kerkhof1, M.J. van Gils1, Z. Euler1, N.L. Verwer1, R.W. Sanders1, H. Schuitemaker1
1

Optimization of the V3 Directed Antibody Response: Strategies to Expose the Epitope


A. Moseri1, T. Subramanyam2, Y. Sagi1, B. Arshava 2, F. Naider2, J. Anglister1
1

Weizmann Institute of Science, Rehovot, Israel; College of Staten Island of the City University of New York, Staten Island, NY, USA
2

Academic Medical Center University of Amsterdam, Amsterdam, Netherlands

Background: V3 directed antibodies with significant breadth and potency have been identified. We studied the structure of V3 peptides in complex with neutralizing antibodies, designed and tested a panel of constrained V3 peptides mimicking the 44752D bound conformation. Our study resulted in identification of an optimally constrained V3 peptide, C4-V3T303C-E322C, which elicits an antibody response that neutralizes SS-1196 and 6535 viral strains. Nevertheless, masking of the V3 loop remains a problem limiting the breadth of V3 directed antibodies. Antibodies such as KD-247 and F425-B4e8, which can neutralize resistant primary isolates such as JR-CSF, recognize a shorter epitope centering the GPGR tip. Methods: The V3 loop can become exposed for neutralization following CD4 binding. CD4M33 is a small and stable CD4-mimic peptide. We have tested the ability of CD4M33 to act in synergy with C4-V3T303C-E322C elicited sera. We are also testing a new methodology of immuno-focusing attempting to focus the antibody response to exposed elements in V3. Optimally constrained peptides at V3T303C-E322C are used for priming followed by additional boost with short V3 peptides representing the F425-B4e8 epitope. Results: CD4M33 was found to act in synergy with C4-V3T303CE322C induced sera in a panel of HIV-1 isolated including SS1196, 6535 and QH0692. Combination-index range from 0.10.5. We have also immunized rabbits with the aim of optimizing the number of priming boosts with the long V3 peptides and the final boosts with the short peptides. Initial binding studies suggest that a single priming step may be optimal. Neutralization studies are currently being performed and will also be presented. Conclusion: The combination of V3 directed vaccine with a preexposure prophylactic administration of a CD4-mimic compounds should be considered as an alternative to other pre-exposure prophylactic drugs. Strategies to expose the V3 loop and expand the breadth of V3 directed antibodies by active immunization should also be pursued.

Background: The HIV-1 envelope glycoprotein (Env) on the viral surface is the target for anti-HIV-1 neutralizing antibodies. In some HIV-1 infected patients broadly neutralizing antibodies (BrNAb) develop that neutralise HIV-1 from different subtypes and therefore are considered to be directed against conserved regions. Understanding of factors involved in BrNAb development will help the rational design of an effective antibody-based vaccine immunogen. Methods: From a previous screening of 299 patients for broadly neutralizing activity (BrNAc), we selected 12 patients with high BrNAc in serum and 10 patients without BrNAc at 3 years after SC. From these patients clonal viruses were obtained within the first year after SC from which gp160 was sequenced and analyzed for length of different regions and number of potential N-linked glycosylation sites (PNGS). Results: There was a trend observed for longer variable regions 1 and 2 (V1V2) in viruses from patients who lacked BrNAc (P=0.16), but there were no significant differences in length of gp160 or different regions of env between the two patient groups. Env from HIV-1 of patients who lacked BrNAc also tended to have a higher number of PNGS (P=0.16), especially in the V1 region, with significantly more PNGS (P=0.044). The number of PNGS in gp41 also tended to be higher in the non-BrNAc group (P=0.10). Conclusion: We hypothesize that the development of BrNAc in HIV-1 infected patients is associated with a more accessible open structure of the viral envelope glycoprotein, which in turn is associated with fewer glycans and shorter variable loops. Testing of epitope specificities in these sera as well as testing of the neutralization sensitivity of these viruses for well defined BrNAbs will help to identify the epitopes on Env that may be responsible for eliciting BrNAc and could help the rational design of an effective antibody-based vaccine immunogen.

POSTERS 104 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.17


Neutralization Potential and Epitope Profiling of Plasma Antibodies from HIV-1 Infected Asian Indians
R. Andrabi1, M. Bala2, R. Kalra1, K. Luthra1
2

Posters

P04.18
The Antibody 2F5 Third ComplementarityDetermining Region of the Heavy Chain (CDRH3) Displays Length Plasticity for Binding and Neutralization
F. Guenaga1, R. Wyatt1
1

All India Institute of Medical Sciences, New Delhi, India; Regional STD Teaching Training and Research Centre, Safdarjang Hospital, New Delhi, India
1

International AIDS Vaccine Initiative, La Jolla, CA, USA

Background: Human immunodeficiency virus type 1 (HIV-1) is the primary cause of AIDS pandemic. Dissecting the specificities of the anti-HIV-1 neutralizing antibodies will assist in identifying targets for an HIV-1 vaccine. Methods: Plasma from 65 drug nave HIV-1 infected patients were tested against a panel of 3 subtype-B (TRO.11, JRFL and RHPA) and 5 subtype-C (Du156.12, ZM53, ZM109F AIIMS201, AIIMS212) tier 1 and tier 2 viruses. Three plasma, found to be broadly neutralizing, were mapped with a set of 211 con-C gp160 overlapping peptides (15mer each). The 50% binding titers were determined by ELISA using consensus-C V3 (35mer), ID loop (19mer) and MPER (24mer) peptides. Statistical analysis was performed using Graph Pad Prism 5. Results: Of the 65 plasma samples, 53 (81.5%) were able to neutralize at least one virus while 12 (18.46%) did not show any neutralization. Only 15 (23%) samples were found to neutralize 50% viruses tested. Clustering analysis revealed that AIIMS201and Du156.12 (clade-C viruses) were the most sensitive while ZM109F, a tier 1b clade-C followed by TRO.11 (clade-B) isolates were the most resistant to antibody neutralization. Epitope specificities of three broadly neutralizing plasma (AIIMS206, AIIMS239 and AIIMS249) with consensus-C gp160 overlapping peptides mapped to V2, V3 and C5 of gp120 and ID loop, MPER and CT of gp41. We did not find any correlation between neutralization capacity and 50% binding titers of anti-V3 (p=0.429), anti-MPER (p=0.683) and anti-ID loop (p=0.680) plasma antibodies. Depletion and competition with V3, MPER and ID-loop peptides showed modest effect on neutralization except for AIIMS239 which showed dependence on MPER directed antibodies. Conclusion: Clustering of neutralization data and phylogenetic analysis suggest the recognition of similar neutralization determinants across different HIV-1 viruses. Identification of epitopes that elicit broadly neutralizing antibodies may serve as effective tool for HIV-1 vaccine design.

Background: The HIV-1 membrane proximal external region (MPER) broadly neutralizing monoclonal antibodies (Mab) 2F5 and 4E10 bind contiguous epitopes in gp41 and are crystallographically characterized in complex with their cognate peptides. The 2F5 structure reveals an extended loop conformation that contrasts with the helix generally adopted by the MPER. Most of the unusually long (22aa) CDRH3 of the 2F5 Mab does not contact the epitope and the CDRH3 possesses a highly hydrophobic apex. Recent data demonstrates that increasing CDRH3 hydrophobicity correlated with neutralization. Here, we aimed at elucidating the involvement of 2F5 CDRH3 length in regards to its capacity to bind or neutralize HIV-1. Methods: We generated 2F5 mutant Mabs that possessed either longer or shorter CDRH3 loops. We next increased hydrophobicity of the length-altered CDRH3s by targeted tryptophan substitutions. We determined Mabs binding affinity constants to epitope and assessed their HIV neutralization activity in pseudovirus assays. Results: The 2F5 CDRH3 tolerated both elongations and reductions of up to four residues, while maintaining nanomolar binding to peptide and HIV neutralization; these activities were enhanced by introducing a V100DW mutation. Substitutions of up to three tryptophans at the apex of the shortened CDRH3 fully reconstituted HIV neutralization activity. Antibody on-rate for the wt MPER peptide correlated positively with neutralization activity. Conclusion: The 2F5 CDRH3 length displayed plasticity for binding its epitope and for neutralization with hydrophobicity of the CDRH3 appearing more critical for neutralization. The positive correlation observed between the on-rate of recognition for the wt MPER peptide and the neutralization activity of the mutant antibodies suggests a model of 2F5 MPER recognition in which the CDRH3 destabilizes the MPER helix to allow the antibody to induce the extended loop peptide-bound conformation observed in the crystal structure.

AIDS Vaccine 2011

105

POSTERS

Posters
P04.19

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.20


Surface Unit but Not Transmembrane Domain Directed Broadly Neutralizing Antibodies Have Differential Potency Against Cell Associated Virus
M. Sagar1, H. Akiyama2, B. Etemad1, N. Ramirez2, I. Freitas1, S. Gummuluru2 Brigham and Womens Hospital / Harvard Medical School, Cambridge, MA, USA; 2Boston University School of Medicine, Boston, MA, USA
1

Identification of Serum SLE-like Auto-Antibodies with Cross Reactivity to Viral Antigens in Patients with HIV-1 Infection
D.C. Alcena1, B. Zheng1, E. Marin1, J. Mattiaco1, M. Keefer1, S. Dewhurst1, X. Jin1, J.J. Kobie1, I. Sanz1
1

University of Rochester School of Medicine and Dentistry, Rochester, NY, USA

Background: A successful HIV-1 vaccine requires the induction of neutralizing antibodies that can protect from infection by a broad range of HIV-1 isolates(BNA). The characteristics of B cells making these antibodies are largely unknown. Several of the BNA are reactive to self-antigens, suggesting a relationship between self-reactivity, B cell tolerance, and the development of BNA. Patients with Systemic Lupus Erythematosis (SLE) have auto-reactive antibodies encoded by VH4.34, also called 9G4 (monoclonal antibody). 9G4 antibody producing cells in healthy individuals are restricted to the naive B cell compartment, yet in SLE patients these 9G4 B cells can undergo germinal center reactions (GC) and become antibody-secreting cells (ASC), leading to elevated serum levels of 9G4. The objective of this study is to examine the association between the presence of 9G4 B cells and BNA activity in HIV-infected patients. Methods: Patients blood samples were obtained. Sera were serial diluted in 9G4 or HIV-1 clade antigen coated 96 well Enzyme-linked Immunosorbant Assay plates and detected using enzyme/substrate reactions. BNA screening was performed using published protocols (ref). Results: Similar to SLE patients (n=35), 9G4 serum antibody titers are significantly elevated in HIV-infected patients (n=112) in comparison to healthy controls (n=28 p<0.05). A fraction of patients (>5%) had significantly greater titers compared to SLE patients. These 9G4 positive antibodies from some HIV-1 patients bind to clade A, B and C HIV-1 Env trimers, and patients with high 9G4+ B cell numbers tend to have BNA to a wide range of HIV strains. Conclusion: Preliminary findings suggest that these 9G4+ B cells in HIV-infected patients may offer an important avenue to study the role of tolerance in the context of induction of a broadly protective anti-HIV-1 antibody response.

Background: Cell free virions are used as the challenge stock in the majority of HIV-1 infection models. Numerous broadly neutralizing antibodies (bNAbs) have been shown to block a diverse array of HIV-1 virions, but it remains unclear if these antibodies exhibit similar potency against cell associated infection, especially dendritic cell trans infection, which potentially plays an important role during mucosal HIV-1 transmission. Methods: Mature DCs were derived from CD14+ monocytes cultured in the presence of IL-4, GM-CSF, and LPS. Mature DCs were exposed to various primary and laboratory adapted HIV1 isolates (MOI=0.2), and washed to remove unbound virus particles. Sensitivity to various bNAbs was examined between cell free and mDC laden HIV-1 in TZM-bl and primary CD4+ T cells. Cell staining was used to examine bNAbs binding mDC and mDC T cell conjugates in the absence of HIV-1. Results: Compared to cell free infection, mDC mediated HIV1 trans infection was significantly less susceptible to gp120 directed bNAbs (VRC01, b12, 2G12). Cell free and mDC associated viruses were equally sensitive to gp41 targeted bNAb (4E10 and 2F5). Broadly NAb, b12, Fab fragment blocked both cell free and mDC-mediated virus infection with equal efficiency. Broadly NAb, 4E10, but not 2G12 bound both mDC alone and mDC T cell conjugate in the absence of HIV-1. Conclusion: Steric hindrance from the tight juxtaposition of HIV1 bearing mDC and target T cell membranes interferes with the ability of gp120 directed bNAbs from blocking mDC-mediated trans infection. On the other hand, gp41 directed bNAbs recognition of host cell lipids allows them to bind mDC prior to the formation of an infectious synapse. Our studies suggest gp120 but not gp41 directed bNAbs may be less potent in preventing HIV-1 spread if transmission or spread from the initial site of infection occurs from a dendritic cell associated source.

POSTERS 106 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.21


Isolation of Potent Neutralizing Monoclonal Antibodies Against V3 Loop from SIV-Infected Macaques
T. Kuwata1, T. Igarashi2, S. Matsushita1
1

Posters

P04.22
Profiles of Neutralizing Antibody Responses in Chronically HIV-1 CRF07_BC Infected Intravenous Drug Users in Western China
X. Hu2, K. Hong2, C. Zhao2, H. Gao1, K.M. Greene1, Y. Zheng2, L. Ren2, Y. Ruan2, D.C. Montefiori1, Y. Shao2 Duke University Medical Center, Durham, NC, USA; 2National Center for AIDS/STD Control and Prevention, Chinese Center, Beijing, China
1

Kumamoto University, Kumamoto, Japan; 2Kyoto University, Kyoto, Japan

Background: Broadly neutralizing monoclonal antibodies from HIV-1-infected patients are an essential but rare tool to understand the mechanism of neutralization against a broad spectrum of HIV-1 strains, including neutralization-resistant strains. Although the humoral immune response to SIV has been studied in the development of vaccine candidates and for exploration of antibodies that efficiently control viral infection, the lack of monoclonal antibodies that can neutralize neutralization-resistant SIV strains is the problem of studying the mechanism of efficient neutralization using the SIV model. Methods: We used phage display method to obtain MAbs against SIV antigens from a SIVsmH635FC-infected rhesus macaque with robust envelope-specific antibody responses. Variable regions of immunoglobulin genes were amplified by rhesus macaquespecific primers and inserted into the phagemid pComb3X to construct phage libraries expressing the Fab fragment. SIVspecific Fab clones were selected by panning with SIV antigen on 96-well plate, and their specificity and neutralizing activities were analyzed. Results: As a result of panning using SIV antigen, many Fab clones specific for SIV Env gp120, gp41 and Gag p27 were obtained. Flow cytometry analysis showed that these Fabs efficiently bound diverse strains of SIVsm/mac, and some of them were cross-reactive with HIV-2. Some of the anti-gp120 Fab clones neutralized the homologous SIVsmH635FC and the genetically divergent SIVmac316, but did not neutralize SIVmac239 and HIV-2GH123. These Fab clones with potent neutralizing activity recognized the same epitope on gp120 including V3 loop. Neutralizing Fab clones specific to the same V3 epitope were also isolated from other SIVsmH635FC-infected macaques. Conclusion: Identification of monoclonal antibodies with potent neutralizing activity against SIV will help to elucidate the mechanisms for inducing broadly neutralizing antibodies in a SIV model.

Background: It is essential to characterize neutralizing antibody (Nab) responses in individuals infected with the diverse HIV1 strains to reveal the potential target for HIV-1 vaccine development. We assess the prevalence, breadth and potency of Nab responses in CRF07_BC chronic infectors(n=100) with infected time around 3 years and compare the neutralization pattern with that of clade B chronically infectors(n=103) with infected time at least 10 years. Methods: 114 plasma samples were collected from intravenous drug users infected with CRF07_BC virus in western China who were ART- nave. The Env pseudovirus-based TZM-bl assay was performed against a panel of tier 2-3 pseudoviruses composed of CRF07_BC(10), clade C(5), B(7), A(4) and CRF01_AE(4) strains and 2 tier1 viruses(SF162.LS and MW965.26). As negative control virus, SVA-MLV positive samples were excluded from further analysis. Results: All 100 plasma samples (excluding 14 SVA-MLV positive samples) neutralized both tier 1 viruses. 53% of samples (n=53) neutralized half of the viruses and 17% of samples (n=17) neutralized more than 80% strains tested. 1% (n=1) neutralized all the viruses and 2% (n=2) neutralized none of the viruses tested. Significant difference of geometric mean ID50 titer (GMT) between intraclade and interclade samples(p<0.001) was observed. CRF07_BC chronically infected subjects showed higher neutralizing activities against subtype-matched viruses than subtype B infectors. However, higher prevalence of broadly crossreactive Nabs response were observed in clade B chronically infected than that of CRF07_BC infection (29% vs 17%). Heatmap analysis against top 6% neutralizers demonstrated that each plasma exhibited unique pattern against the spectrum of viruses. Conclusion: We detected relatively high broadly cross-reactive neutralization activities in 17% plasma of CRF07_BC infectors tested. Further epitope specificity dissection of these broadly cross-reactive Nab samples will provide useful insights for rational HIV-1 vaccine design.

AIDS Vaccine 2011

107

POSTERS

Posters
P04.23

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.24


Detection of CD4-Binding Site Antibodies in Three HIV-1 Seroconverter Cohorts
R.M. Lynch1, M.K. Louder1, L. Tran1, Z. Yang1, G. Tomaras2, M. Cohen3, E.S. Gray4, S. Sibeko5, S.A. Karim5, Z. Euler6, G.J. Nabel1, H. Schuitemaker6, D.C. Montefiori2, L. Morris4, B.F. Haynes2, J. Mascola1
1 National Institutes of Health, Bethesda, MD, USA; 2Duke University Medical Center, Durham, NC, USA; 3University of North Carolina School of Medicine, Chapel Hill, NC, USA; 4 National Institute for Communicable Diseases, Sandringham, South Africa; 5University of KwaZulu-Natal, Durban, South Africa; 6Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands

Plasmacytoid Dendritic Cells Infection by HIV-1 Is Inhibited by Neutralizing Antibodies Without Interfering with IFN- Production
A. Lederle , J. Penichon , G. Laumond , T. Decoville , S. Schmidt1, V. Holl2, C. Moog1
1 1 1 1 1 2

UMR INSERM/UdS U748, Strasbourg, France, Metropolitan; Covance CLS SA, Meyrin, Switzerland

Background: Plasmacytoid dendritic cells (PDC) are involved in innate and adaptive immunity, and produce large amounts of antiviral type I interferons. These cells are productively infected by HIV-1 in vitro and in vivo, and several studies revealed a decrease in circulating PDC number, correlated with an increased plasma viral load. We have previously showed an FcRs-mediated inhibition of myeloid DC infection by neutralizing (NAbs) as well as nonneutralizing inhibitory (NNIAbs) antibodies. The aim of this study is to analyze the mechanism of inhibition of PDC infection by Ab. Methods: Neutralization assay was performed with primary human healthy PDC or GEN2.2 PDC cell line in the presence of serial antibodies concentrations. The percentage of infection was quantified by flow cytometry using intracellular p24 viral antigen staining and IFN- production was measured by CBA flex. Results: We showed that NAbs strongly inhibited R5-HIV-1 replication in GEN2.2 cell lines. Although they expressed FcRII, we have not detected an inhibitory activity of NNIAbs. We also found an efficient inhibition of primary PDC infection by NAbs, but not with NNIAbs, with similar activities that those measured with GEN2.2 PDC cell line. Interestingly, a strong level of IFN- production was maintained in primary PDC infected by HIV, even when the HIV-1 replication was inhibited by NAbs. Conclusion: We showed that 1) HIV-1 infection of PDC is inhibited by NAbs, 2) FcRIIa seems not to be involved in this inhibitory process, and 3) the inhibition of HIV-1 replication by NAbs do not preclude IFN- production by PDC. Overall these results suggest that HIV-1-specific NAbs induced by an efficient vaccine should prevent PDC infection, without interfering with the release of IFN- and thus the anti-viral innate immune response.

Background: Broadly reactive neutralizing antibodies to the CD4 binding site (CD4bs) of gp120 have been detected in some chronically HIV-1 infected individuals. However, little is known about the prevalence and evolution of this antibody response from the beginning of infection. Methods: Here, we analyzed 108 sera, derived from three cohorts of HIV-1 seroconverters, for binding to a panel of gp120 core proteins and their corresponding CD4bs mutants. More specific CD4 binding site epitope binding was assessed using the resurfaced stabilized core (RSC3) or outer domain (OD4.0) proteins that preferentially bind the CD4bs antibodies b12, b13, m18, VRC01, 02 and 03. Results: Among sera collected between 2-5 years post-infection, over 80% contained anti-CD4bs antibodies, and over 50% contained antibodies reactive with RSC3 and/or OD4.0. Five of the latter subjects were followed longitudinally, and these CD4bs antibodies were found to arise between weeks 12-72 post-infection. However, despite the presence of these binding antibodies, CD4bs neutralizing activity was not detectable by 96 weeks post-infection. Conclusion: Thus, binding antibodies with profiles similar to b12, b13, m18, VRC01, 02 and 03 can develop within the first year of infection and are present in a majority of HIV-1 infected subjects from three cohorts across the globe. However, within the first two years of infection, these binding antibodies do not contribute to the neutralizing activity of the sera. These data suggest that although the CD4bs is immunogenic, antibodies directed against it may require epitope refinement or maturation over time in order to neutralize the viral spike. Further study of how these antibodies develop and evolve over time may provide clues for immunization strategies aimed at eliciting similar antibody responses.

POSTERS 108 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.25


Elicitation of Neutralising Antibodies Using Patient-Derived gp140 Immunogens from a LongTerm Non-Progressor
E.J. Bowles1, J. Parker1, L. Heyndrickx2, B. Grevstad3, M. Jansson4, G. Vanham2, G. Stewart-Jones1
1

Posters

P04.26
Extensive Profile of the Anti-HIV Inhibitory Activity of Well-Known Monoclonal Antibodies, Possible Correlates for In Vivo Protection
M.E. Biedma1, S. Schmidt1, T. Decoville1, G. Laumond1, V. Holl2, C. Moog1
1 2

Oxford University, Oxford, United Kingdom (Great Britain); Institute of Tropical Medicine, Antwerp, Belgium; 3Statens Serum Institut, Copenhagen, Denmark; 4Lund University, Lund, Sweden
2

UMR INSERM/UdS U748, Strasbourg, France, Metropolitan; Covance CLS SA, Meytin, Switzerland

Background: Broadly neutralising antibodies (bNAbs), that crossreact with diverse HIV-1 subtypes, develop in some individuals during chronic infection. Immunisation strategies that utilise patient-derived envs may result in improved bNAb development. We studied the env sequence evolution in ITM-1, a long-term non-progressor patient with bNAbs. We selected two env vaccine candidates from the phylogeny and tested their immunogenicity in rabbits. Methods: HIV-1 env was extracted, amplified and sequenced from 30 plasma samples collected over 11 years from ITM-1 and 7 plasma samples from his mother. Sequence evolution was analysed following alignment and phylogeny reconstruction. The earliest env, ITM_1_4, and the predicted ancestral env, ITM_anc, were selected for use as immunogens. gp140 trimeric proteins were expressed in 293T cells and purified using metal-affinity, lectin and FPLC. Rabbits were immunised at weeks 0, 2, 4 and 8 with 100ug trimer adjuvanted with CAF01. Sera collected at weeks 0, 2, 4, 8, 12 and 14 were screened in gp120 ELISA and TZM-bl and PBMC neutralisation assays. Results: ITM-1 and Mother env sequences diverged into distinct lineages following transmission. We observed substantial variable loop expansion, variations in glycosylation and an accumulation of positively-selected substitutions over time in ITM-1 sequences. We selected two early envs from the phylogeny for immunisation studies with trimeric gp140 protein. All rabbits generated a gp120specific IgG response 2 weeks after the first dose. Titres were boosted after each subsequent immunisation. Sera from ITM_1_4 immunised rabbits were able to neutralise Tier 1 pseudoviruses SF162 and BX08. Neutralisation was also detected in ITM_anc immunised rabbits, though the titres were lower. Conclusion: Early isolates from patients who develop bNAbs can be used to design gp140 immunogens which may induce greater neutralisation breadth in HIV-1 vaccination studies. A clearer understanding of what drives bNAb development will further inform vaccine design.

Background: Fc receptors (FcRs) are differentially expressed on a variety of cells. Fc-FcR interactions play a critical role in the biological function of antibody and can mediate several innate immune mechanisms. Particularly various inhibitory activities of antibodies against HIV detected in vitro may impact on sterilizing protection in vivo. Therefore, the different immune activities mediated by well known HIV-specific monoclonal Ab were evaluated using different effector cells. Methods: Neutralizing activities (using TZMbl and PBMC assay) Fc-mediated inhibitory activities (on macrophages and dendritic cells), ADCC (on primary NK effectors and HIV infected target cells) and capture assay of primary isolates were carried out for well known anti-HIV specific Abs. Results: We show that these HIV-1 specific antibodies displayed various inhibitory activities: These activities were ranked differently depending on the Ab, one being more efficient for neutralization, the other for Fc-mediated inhibition, or ADCC etc. Conclusion: This study gives a general pattern of the inhibitory activities of the well known HIV specific antibodies. The inhibitory profile of each Ab will help to define the key inhibitory activities contributing to in vivo protection. Specific combination of Abs may be particularly favourable by cumulating inhibitory activities leading to efficient protection from HIV infection.

AIDS Vaccine 2011

109

POSTERS

Posters
P04.27

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.28


Neutralisation Activity of HIV-1 Clade C- and CRF02_AG-Infected Serum/Plasma: Some Viruses Are Not as Resistant as Previously Thought
R.A. Jacob1, F. Abrahams1, M. Tongo2, P. Roux3, E.M. Ngole4, W.A. Burgers2, J.R. Dorfman1
1

Combined HIV-1 Neutralization Coverage by VRC01, PG9, and PG16


N.A. Doria-Rose1, X. Wu1, Z. Yang1, M.K. Louder1, K. McKee1, S. ODell1, S. Schmidt1, W.C. Koff2, M.S. Seaman3, R. Bailer1, D.R. Burton4, J.R. Mascola1
1

National Institutes of Health, Bethesda, MD, USA; International AIDS Vaccine Initiative, New York, NY, USA; 3 Beth Israel Deaconess Medical Center, Boston, MA, USA; 4The Scripps Research Institute, La Jolla, CA, USA
2

Background: Neutralizing antibodies (NAb) are considered a critical component of an effective vaccine for HIV-1 and have been shown to provide sterilizing protection in animal models. Three recently described monoclonal antibodies, VRC01, PG9, and PG16, have extraordinary potency and breadth of neutralization. These human mAbs are potential models for immunogen design, and are of interest for clinical use to prevent HIV-1 infection. Therefore, we analyzed the potential total coverage of HIV-1 isolates provided by combinations of these antibodies. Methods: Neutralization titers were determined for each antibody against a panel of 199 HIV-1 Env pseudovirus isolates using TZMbl target cells. Isolates included clades A, B, C, D, and G and CRFs AC, AD, AE, AG, ACD, BC, and CD. IC50 and IC80 titers were used for combination analysis; sensitivity or resistance was determined using cutoffs of 50 ug/ml or 1 ug/ml. Results: Individually, VRC01 neutralized 90.4% of isolates with IC50 < 50, PG9 neutralized 78.4%, and PG16, 73.9%. At the most stringent cutoff we considered, IC80 < 1, the individual NAbs neutralized 51.8%, 45.2%, and 42.2% respectively. Together, VRC01 + PG9 neutralized 97.0%, VRC01+PG16 neutralized 96.5% and PG9 + PG16 neutralized 80.4% at an IC50 < 50. Using an IC80 < 1, the values were 69.9%, 70.9% and 50.2% respectively. Only 6 out of the 199 isolates were fully resistant to both VRC01 and PG9. Resistance to VRC01 was independent of resistance to PG9 or PG16. IC50 titers for VRC01 did not differ between isolates that were resistant or sensitive to PG9 or PG16 and vice versa. Conclusion: The combination of VRC01 with PG9 or PG16 provides neutralization of up to 97% of global HIV-1 strains. These combinations could be very useful clinically in therapeutic or prophylactic applications.

International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa; 2Division of Medical Virology, University of Cape Town, Cape Town, South Africa; 3School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa; 4Institut de Recherche Medicale et dEtude des Plantes Medicinales, Yaounde, Cameroon
Background: Significant strides in understanding neutralisation breadth of HIV-1 have been made recently. In order to develop a global vaccine, it remains important to understand the neutralisation of the wide diversity of viruses that circulate globally. Methods: We developed a panel of 24 HIV-1 pseudoviruses selected for resistance to neutralisation, diversity by clade, geographic origin and sequence. We tested this panel using 70 sera from an ARV-naive cohort infected with HIV-1 (presumed subtype C) for >1yr and 10 plasma samples from a Cameroonian blood bank that were infected with typed CRF02_AG-related viruses and were selected to be comparable in neutralisation capacity to our South African samples. Results: The neutralising responses of the subtype C sera were broad, with 8/70 sera tested neutralising 18/24 target pseudoviruses at ID50>100, and 12/70 neutralizing 16/24 target pseudoviruses. We found that most clade A and some CRF02_AG target pseudoviruses tended to be the least sensitive to neutralisation by our sera from presumed clade C-infected individuals, followed by Clade B and then clade C viruses. Surprisingly, and in contrast to previous reports, plasma from clade CRF02_AG infected donors neutralised several of the CRF02_AG viruses well, including two Tier 3 (251-18 and 33-7) viruses that were previously described as highly resistant. Conclusion: Some but not all CRF02_AG-related viruses previously described as tier 3 are sensitive to neutralisation by CRF02_AGdervied plasma, even though all are previously reported as highly resistant. Further work to identify which viruses are intrinsically resistant to neutralisation is needed. Three viruses were identified that were most resistant to both South African and Cameroonian samples, which are now major targets in our monoclonal antibody project. Focus should be placed on those viruses that are intriniscally resistant to neutralisation in order to develop an HIV-1 vaccine that does not select for such viruses.

POSTERS 110 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.29


Early Autologous Neutralising Epitopes in C3V4 and 14+V5 Are Readily Adsorbed with Monomeric gp120
C.K. Wibmer1, J.N. Bhiman1, P.L. Moore1, N.C. Ranchobe1, S.A. Karim2, L. Morris1, E.S. Gray1
1

Posters

P04.30
Features and Diversity of Quaternary Neutralizing Epitopes (QNEs) in HIV-1 Env that Mediate Potent Viral Neutralization
C. Krachmarov1, W. Honnen1, Z. Lai1, M. Gorny2, S. Zolla-Pazner2, J. Robinson3, A. Pinter1
1

National Institute for Communicable Diseases, Johannesburg, South Africa; 2Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa

The Public Health Research Institute, UMDNJ, Newark, NJ, USA; 2New York University, Langone School of Medicine, New York, NY, USA; 3Tulane University Medical Center, New Orleans, LA, USA

Background: The earliest neutralising antibody responses target epitopes predominantly in variable loop regions of gp120. We have previously shown that the C3 region and V4 loop often form a discontinuous epitope for these antibodies in acute subtype C infection. This work identified another discontinuous neutralisation epitope comprising the 14 sheet in C3 and adjacent V5 loop. Since C3-V4 and 14+V5 exist entirely within the structurally conserved outer domain of gp120, it was hypothesised that autologous antibodies targeting these regions could be adsorbed with monomeric gp120. Methods: Recombinant gp120s from the transmitted/founder virus of four HIV-1 infected individuals were expressed in 293T cells and purified by lectin affinity and ion exchange chromatography. Proteins were conjugated to tosyl-activated magnetic beads and used to adsorb the neutralisation activity in autologous plasma from a time point with peak titres for the epitope of interest. Chimeric viruses engineered by overlapping PCR were tested for neutralisation sensitivity to these plasmas, and recombinant chimeric proteins were expressed for adsorption studies. Potential escape mutations were introduced via sitedirected mutagenesis. Results: Similar to antibodies targeting C3-V4, neutralising antibodies targeting the 14+V5 region of gp120 developed to high titre during acute infection. In both instances these antibodies could be adsorbed with monomeric gp120; however this adsorption did not often account for the total neutralisation activity at a particular time point. These residual titres were shown to be the result of concurrent antibodies against quaternary epitopes in the V1-V2 domain. Escape from C3-V4 and 14+V5 antibody specificities were due to changes in charge or glycosylation within the epitope, with the exception of one individual that escaped C3-V4 neutralising antibodies through the acquisition of a glycan in V1. Conclusion: Collectively this data identified two key immunogenic regions on monomeric gp120 often targeted by early neutralising antibody responses during acute subtype C infection.

Background: MAbs specific for QNEs that potently neutralize HIV-1 have been isolated from infected humans and monkeys. A common feature shared by these antibodies is a dependence on specific residues in both the V2 and V3 domains of gp120. In this study we compared the V2 and V3 determinants of human mAb 2909 with those of 11 mAbs from infected macaques. Methods: Mutant HIV-1 Envs were generated by site-directed mutagenesis. Neutralizations were performed via single-cycle infectivity assays in U87-CD5-CCR5 cells, using luciferaseexpressing virions pseudotyped with the Env proteins. Flow Cytometry was used to analyze the formation of QNEs between subunits of the trimer. Results: The fine specificity of the monkey mAbs for both V2 and V3 regions differed from that of 2909 and from each other. While the limited breadth of 2909 was due to the requirement of a single substitution at position 160, the monkey mAbs were also dependent on substitutions at positions 167169, and, in most cases, required R315 at the tip of the V3 loop. Positions 167 and 169 were also found to significantly affect masking of standard epitopes in the V3 and CD4-binding domains. Flow cytometry of co-expressed Envs containing either the V2 or V3 components suggested that these epitopes were expressed on single gp120 molecules, and not on neighboring protomers. Conclusion: The different epitope specificities of 2909 and the monkey mAbs indicate that specificity and breadth are strongly influenced by the sequence of the infecting virus. Comparisons with QNEs recognized by the PG9/PG16 mAbs and CAP256 plasma suggested that these overlapped, adding to mounting evidence that the N terminal region of the V2 domain is a component of such neutralization targets. The unprecedented neutralization potency of QNE-specific antibodies argues that these represent extremely sensitive neutralization targets, and could be important targets for HIV-1 vaccines.

AIDS Vaccine 2011

111

POSTERS

Posters
P04.31

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.32


Antibody-Guided Exploration of V3 Exposure on HIV Subtype C
S. Manhas1, B. Clark1, S. Zolla-Pazner2, M.K. Gorny2, R. Pantophlet1 Simon Fraser University, Vancouver, Canada; 2New York University, New York, NY, USA
1

Sequential DNA Prime-Protein Boost Is More Effective in Eliciting HIV-1 Env-Specific Antibody Than Simultaneous Delivery of DNA + Protein Components
Y. Chen1, S. Wang1, S. Lu1
1

University Massachusetts Medical School, Worcester, MA, USA

Background: Our recent work has demonstrated that DNA prime-protein boost immunization is more effective than protein alone approach in eliciting higher overall antibody responses against HIV-1 Env and more importantly broader neutralizing antibodies against primary HIV-1 across different clades. At the same time, studies in the non-HIV vaccine field have suggested that co-delivery of DNA and protein may suppress T cell immune responses. Methods: In the current study, we conducted mouse experiments to identify whether simultaneous delivery of HIV-1 gp120-expressing DNA vaccine and recombinant gp120 protein will be equally effective as sequential DNA prime-protein boost in eliciting Envspecific antibody responses. Mice were immunized twice by one of the following designs: 1) DNA-DNA, 2) protein-protein, 3) DNA-protein; 4) DNA/protein-DNA/protein. Serum gp120-specific antibody titers were measured at the end of two immunizations. Results: Our results showed that the protein alone approach was the least effective. While DNA/protein-DNA/protein was more effective than the protein alone approach in eliciting slightly higher antibodies, it was much less effective than the DNA-protein approach. Interestingly, DNA/protein-DNA/protein approach elicited lower antibody responses than the DNA-DNA approach, supporting the previous claim that adding a protein component to the DNA vaccine (coding for the same antigen) can be suppressive to the original immune responses elicited by a DNA alone vaccine. We further identified that such suppression is dose dependent, i.e. higher doses of co-delivered DNA/protein vaccines lead to lower antibody responses. Conclusion: The above results indicated that DNA prime plays a critical role in immune activation and this finding will have important implications on the future design of prime-boost HIV vaccines. This work was supported in part by NIH grants AI065250, AI082274 and AI082676.

Background: Elucidating the exposure of antibody epitopes on HIV Env is important for vaccine immunogen design. While relatively much effort has been devoted to exploring accessibility of the V3 region on subtype B viruses, V3 exposure on subtype C viruses has yet to be fully understood. V3 is relatively more variable among subtype B than subtype C. However, the C3 region in subtype C exhibits greater sequence variability than in subtype B, suggesting different immunological pressures and possibly unique epitope exposure between these two subtypes. Investigating potential subtype-specific differences in V3 exposure may provide greater understanding of Env oligomeric structure and be insightful to immunogen design efforts. Methods: To assess the accessibility of V3 on subtype C viruses, anti-V3 monoclonal antibodies (MAbs) B4e8, 2219, and 268-D, isolated from subtype B infected individuals, and anti-V3 MAb 2557, isolated from a CRF02_AG infected individual, were tested for their ability to neutralize a panel of 10 subtype C primary isolates using a luciferase-based single-round pseudovirus assay. Results: Of the 10 subtype C isolates tested, 6 were not neutralized significantly by any of the 4 anti-V3 MAbs (IC50 <50% at 50 g/ml). Unexpectedly however, the remaining 4 viruses (ZM53M, ZM249M, ZM233M and ZM197M) were neutralized with increasing potency at low antibody concentration (IC50 >50% at 2 g/ml). Conclusion: The results suggest that in some subtype C viruses V3 might be more accessible to antibody than previously appreciated. To determine if neutralization of ZM53M, ZM249M, ZM233M and ZM197M at low antibody concentration is V3 specific, studies are ongoing to explore the sensitivity of these viruses to non-V3 MAbs. Studies are also ongoing to investigate the influence of the C3 region on V3 exposure by generating chimeras of V3 neutralization-sensitive and -resistant subtype C viruses. These chimeric viruses will be tested for neutralization sensitivity to V3 and non-V3 MAbs.

POSTERS 112 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.33


Epitope Specificity and Envelope Diversity in an Elite Neutralizer from the San Francisco Bay Area
S.M. ORourke4, J. Robinson1, M.A. Conant1, B. Schweighardt2, P. Phung2, K. Limoli2, T. Wrin2, K. Higgins3, F. Sinangil3, P. Berman4
1 The Conant Foundation, San Francisco, CA, USA; 2Monogram Biosciences, South San Francisco, CA, USA; 3Global Solutions for Infectious Diseases, South San Francisco, CA, USA; 4 University of California, Santa Cruz, Santa Cruz, CA, USA

Posters

P04.34
Mapping the Specificity of Broadly Neutralizing Antibodies from Thai Elite Neutralizers by Analysis of crf A/E Virus Quasi-Species
S.M. ORourke4, R. Sutthent1, G. Tatsuno4, B. To4, P. Phung2, K. Limoli2, K. Higgins3, T. Wrin2, F. Sinangil3, P. Berman4 Microbiology Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; 2Monogram Biosciences, South San Francisco, CA, USA; 3Global Solutions for Infectious Diseases, South San Francisco, CA, USA; 4 University of California, Santa Cruz, Santa Cruz, CA, USA
1

Background: In this report we describe an elite neutralizer (EN) with unusually potent broadly neutralizing antibodies. We have used swarm analysis of viral quasi-species to define the populations of antibodies responsible for this broad neutralizing activity, and to understand the characteristics of the envelope proteins that stimulated this unusual immune response. Methods: Sera from 17 volunteers were screened for broadly neutralizing antibodies (bNAbs) with pseudotype viruses from 5 different clades. Envelope genes were cloned from GSID001 plasma, and 11 with high infectivity were sequenced and tested for neutralization sensitivity/resistance. Additional neutralization studies were carried out with pairs of viruses from other donors. Results: Sera from GSID001 potently neutralized 23 of 24 viruses in a panel of international isolates (GMT=1179). While most of the autologous clones from subject GSID001 were resistant to neutralization, clone 029 was sensitive to neutralization. Sequence analysis identified 60 amino acid differences between clone 029 and neutralization resistant clone 012. Site directed mutagenesis was used to localize residues responsible for this difference. Testing of viral envelope clones from other donors yielded an additional 7 pairs of neutralization sensitive/resistant envelopes (5 clade B and 2 CRF A/E from Thailand) appropriate for epitope mapping. For example these studies showed that deletion of a glycosylation site at position 196 (N196H) in the V2 domain markedly enhanced neutralization sensitivity by GSID001 as well as another EN, Z23. Studies in progress have localized additional epitopes responsible for the broadly neutralizing activity. Conclusion: The antibody response in GSID001 sera is an example of the type of broadly neutralizing immune response that we wish to elicit with a candidate HIV vaccine. Understanding the specificity of the antibody populations responsible for this broad neutralizing activity, and the antigenic structure of the envelope proteins that stimulated this unusual immune response, should allow us to build improved vaccine antigens.

Background: A major goal of HIV vaccine development is to identify immunogens that elicit broadly neutralizing antibodies (bNAbs) similar those found in rare HIV-infected individuals termed elite neutralizers. Although a few epitopes recognized by bNAbs have been defined for individuals infected with clade A, B, and C viruses, they have not been described for clade E (crf A/E) viruses. In this report we analyzed the specificity of neutralizing antibodies from Thai elite neutralizers using viruses collected from a cohort of injection drug users from Bangkok. Methods: Envelope gene quasi-species were amplified from 30 individuals, and pseudotype viruses were evaluated for sensitivity and resistance to antibodies from 3 Thai elite neutralizers. Pairs of neutralization sensitive and resistant viruses were identified and amino acids responsible for neutralization sensitivity were localized by site directed mutagenesis. Results: Analysis of mutations in Thai viruses that conferred neutralization sensitivity and resistance appeared to correspond to epitopes recognized by bNAbs. In contrast, mutations in North American viruses identified by this approach appeared to correspond to mutations that induced conformational changes affecting multiple epitopes. Moreover we found that different populations of neutralizing antibodies were present in different elite neutralizers. In some cases different antibody populations from different elite neutralizer sera could be mapped with virus quasi-species from a single patient. Mutations that conferred neutralization sensitivity/resistance included a unique site in the V1 domain, and a conserved residue in the V3 domain. Other sites recognized by different individuals in needle sharing transmission linkage groups are currently under investigation. Conclusion: These studies identify unique amino acids that affect the binding of bNAbs from Thai elite neutralizers. Envelopes with these mutations potentially represent a new source of improved vaccine immunogens. It will be interesting to determine whether antibodies to the sites identified might represent potential correlates of protection in the RV144 vaccine trial.

AIDS Vaccine 2011

113

POSTERS

Posters
P04.35 LB

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.36 LB


Structures of HIV-1 Quaternary-StructurePreferring Antibodies, CH04 And PG9, Show Conserved Structural Elements Within a Generallly Flexible CDR H3
R.K. Louder1, J. McLellan1, M. Pancera1, S.D. Schmidt1, Y. Yang1, B. Zhang1, S. Phogat2, M. Bonsignori3, K. Hwang3, H. Liao3, X. Chen3, M.A. Moody3, B.F. Haynes3, J.R. Mascola1, P.D. Kwong1 National Institutes of Health, Bethesda, MD, USA; International AIDS Vaccine Initiative, Brooklyn, NY, USA; 3 Duke University School of Medicine, Durham, NC, USA
1 2

Neutralization of Plasma vs. Antibodies: The HJ16, HGN194 and HK20 Comparisons
S.S. Balla-Jhagjhoorsingh1, B. Willems1, L. Heyndrickx1, K. Vereecken1, D. Corti2, A. Lanzavecchia2, D. Davis3, G. Vanham1 Institute of Tropical Medicine, Antwerp, Belgium; 2Institute for Research in Biomedicine, Bellinzona, Switzerland; 3 Biomedical Primate Research Centre, Rijswijk, Netherlands
1

Background: Several new human antibodies with a neutralizing potential across different subtypes have recently been described. Three mAbs, HJ16, HGN194 and HK20, were obtained from cells from three Sub-Saharan African women within the HIV-1 cohort of the Institute of Tropical Medicine. Methods: Polyclonal plasma from these HIV-1 infected patients was compared to mAbs obtained from their memory B-cells. PBMC based neutralization assays with varying incubation (1h vs 24h), adsorption (1-2 h vs 24 h) and culture phases (7 or 14 days) were performed. We compared these PBMC assays with cell line based assays using TZMbl and GHOST cells. The role of primary replicating virus versus non-replicating pseudovirus was considered using a panel of tier 1 and 2 strains. Results: The results clearly demonstrate that patient selection was highly dependent on the neutralization assay. Although the cross-neutralizing properties of the isolated Abs showed considerable variation with the neutralization assay format, all assays indicate that neutralizing Abs to other epitopes of the HIV-1 envelope than those targeted by the obtained mAbs are present in the plasma. Conclusion: The present study indicates that different neutralization assays yield different results and it is still unclear which one is most predictive of in vivo neutralizing activity. Moreover, the strong profiles in the patients plasma were not solely due to antibodies represented by the newly isolated mAbs. This better understanding of in vitro neutralization characterizations of patient plasma and Abs will hopefully lead to more effective ways of discovering new Abs that ultimately can be used for HIV-1 immunogen design and subsequent vaccine development.

Background: Broadly neutralizing quaternary-structure-preferring antibodies, such as PG9, PG16, and more recently CH01-CH04, have been isolated from HIV-infected patients. These antibodies bind to a glycosylated epitope comprised of the second and third variable loops. They are highly affinity-matured and have long heavy chain 3rd complementarity-determining regions (CDR H3) of 24 to 28 amino acids in length. Although they neutralize a broad range of viruses, they can only bind monomeric gp120s from select strains, indicating that they preferentially recognize the oligomeric conformation of the HIV-1 spike. Methods: To investigate paratope elements of recognition for the quaternary-structure-preferring antibodies, we obtained crystals of CH04, CH04H-light (CH04 heavy chain but a different light chain), and PG9 antigen-binding fragments (Fab) that diffracted X-rays to 1.9 , 1.8 and ~3 , respectively. Results: Here we show that the CDR H3 of CH04, CH04-light and PG9 is highly flexible and can adopt different conformations depending on the crystal lattice. However, in each structure where the CDR H3 is ordered, a two-stranded antiparallel -sheet element is conserved towards the tip of the CDR H3. This structural element was also observed in the PG16 Fab structure. Conclusion: The unusually long CDR H3 of the quaternarystructure-preferring antibodies appears to be highly flexible and might adopt different conformations upon binding to the quaternary-structure-preferring epitope. Nevertheless, a conserved -sheet structural element appears to be required to bind to the epitope.

POSTERS 114 AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.37 LB


Structures of Reverted and Mature VRC01-Like Antibodies Illustrate the Evolution Required to Generate Effective HIV-1 Neutralizing Antibodies
M. Joyce1, T. Zhou1, M. Pancera1, S. Moquin1, X. Wu1, B. Zhang1, N.J. Lu1, J. Zhu1, X. Chen1, I. Georgiev1, L. Shapiro1, J.R. Mascola1, G.J. Nabel1, P.D. Kwong1
1

Posters

P04.38 LB
Identification of Minimal Antigenic Domains In HIV-1 Envelope Glycoprotein Recognized by Broadly Neutralizing Monoclonal Antibodies VRC01 And PG9
H. Wang1, X. Shi1, C. Yao1, T. Zuo1, L. Zhang1
1

Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA

AIDS Research Center, School of Medicine, Tsinghua University, Beijing, China

Background: A group of highly effective neutralizing antibodies (VRC01/VRC03/VRC-PG04/VRC-CH31), which target the site of CD4 binding on HIV-1 gp120 have recently been identified. Intriguingly, antibody-gp120 complex structures show that recognition occurs in a structurally conserved manner. Although there is some degree of commonality in the VRC01-like antibody germline genes, the final matured antibodies display high sequence diversity. To understand the structural evolution of VRC01like antibodies from initial recombinant to mature neutralizing antibody, we will characterize the free/unbound structures of (i) reverted germline antibodies, (ii) phylogenetically inferred intermediate antibodies, (iii) intermediate antibodies identified by deep sequencing, and (iv) mature neutralizing antibodies. Methods: Intermediate antibodies were identified by 454 pyrosequencing and functional genomics. Cross-donor phylogenetic analysis was used to identify inferred intermediates. Antibodies were produced by expression in 293F cells and Fab domains were generated by typical methods and crystallized by vapor diffusion. Results: Structures of several mature unbound Fab domains have been determined as well as the unbound V-gene reverted Fabs from VRC03 and VRC-PG04. Comparison of gp120-bound and unbound mature antibody structures reveals minimal structural changes upon gp120 binding, suggesting that mature VRC01-like antibodies utilize a lock-and-key mechanism in their recognition. Meanwhile, the reverted VRC03 and VRC-PG04 structures reveal that the CDR L1, CDR H1, H3, and framework H1 regions display structural changes when compared to the mature antibody structures while the CDR L3, CDR H2 and framework H3 region, including the critical Arg 71 remain structurally unchanged. Conclusion: Structurally conserved residues appear to be important in initial HIV-1 recognition while regions displaying structural changes are likely important in the antibody affinity maturation process. Interestingly, structurally conserved regions involve hydrophobic interactions; a precise energetic landscape of antibody-gp120 interactions combined with the structural data will allow an in-depth understanding of the VRC01-like antibody maturation process.

Background: VRC01 and PG9, identified by far as the two most potent human broadly neutralizing monoclonal antibodies (bnmAb) against HIV-1, are able to neutralize large assay of primary viruses of diverse genetic and geographic origins. Identification and characterization of the antigenic domains recognized by the two bnmAbs and their germline precursors may therefore provide valuable insights into better understanding of the mechanism of neutralization and the rational design of vaccines. Methods: Here, we report the development of an efficient mapping technique for minimal antigenic domains based on the combinatorial antigen library of HIV-1 envelope glycoprotein displayed on the surface of the yeast Saccharomyces cerevisiae. In this technique, positive yeast clones recognized by the two bnmAbs were identified and obtained by fluorescence-activated cell sorting (FACS) followed by sequencing and structural analysis. Results: Using this technique, we have identified several protein fragments reactive to VRC01 and PG9 that are variable in length and overlapping in nature. Although each protein fragmentantibody complex demonstrates a distinct profile of fluorescence intensity, those recognized by VRC01 invariably contain the V3, V4 and V5 regions while those of PG9 contain the V1 and V2 regions of gp120. These protein fragments very likely resemble the minimal antigenic domains recognized, as they contain neither the V1 and V2 regions shown to interact with VRC01 nor the V3 region with PG9 previously reported in structural and mutagenesis studies. Our results suggest that the major energetic binding residues for VRC01 or PG9 must be confined within the selected fragments, although detailed mapping of these residues requires further investigation. Conclusion: We believe the novel mapping technique and minimal antigenic domains identified here are unprecedented. Application of this technique to study other bnmAb and their germline precursors will improve our understanding of protective immunity and guide rational design of vaccines.

AIDS Vaccine 2011

115

POSTERS

Posters
P04.39 LB

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.40 LB


Structural Basis for VRC01-Like-Antibody Recognition of Variable Loops that Surround the Site of CD4 Attachment on HIV-1 gp120
T. Zhou1, X. Wu1, B. Zhang1, S. Moquin1, J. Zhu1, I. Georgiev1, M. Bonsignori2, J.A. Crump3, S.H. Kapiga4, S.E. Noel5, B.F. Haynes2, M. Simek6, D.R. Burton7, W.C. Koff6, L. Shapiro8, J.R. Mascola1, P.D. Kwong1
1 NIAID/NIH, Bethesda, MD, USA; 2The Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA; 3Dept. of Medicine and Dept. of Pathology, Duke University Medical Center, Durham, NC, USA; 4Kilimanjaro Reproductive Health Programme, Moshi, United Republic of Tanzania; 5Tumaini University, Moshi, United Republic of Tanzania; 6International AIDS Vaccine Initiative (IAVI), New York, NY, USA; 7The Scripps Research Institute, La Jolla, CA, USA; 8Dept. of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA

Isolation of CD4-Binding Site and V2/ V3 Conformational (Quaternary) Broadly Neutralizing Antibodies from the Same HIV-1 Infected African Subject
M. Bonsignori , X. Wu , M.A. Moody , H. Liao , K. Hwang , J.A. Crump3, S.H. Capiga4, N.E. Sam5, G.D. Tomaras1, X. Chen1, C. Tsao1, S.M. Alam1, G.J. Nabel2, P.D. Kwong2, L. Morris6, D. Montefiori7, J.R. Mascola2, B.F. Haynes1
1 2 1 1 1

Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; 2Vaccine Research Center, NIH, Bethesda, MD, USA; 3Department of Medicine, Duke University Medical Center, Durham, NC, USA; 4London School of Hygiene and Tropical Medicine, London, United Kingdom (Great Britain); 5Kilimanjaro Christian Medical Centre, Moshi, United Republic of Tanzania; 6National Institute for Communicable Diseases, Johannesburg, South Africa; 7 Department of Surgery, Duke University Medical Center, Durham, NC, USA
1

Background: Recent studies have demonstrated the production of broadly neutralizing antibodies (bNAbs) in ~20% of chronically infected subjects and remarkable bNAb magnitude and breadth in the top 1-3% (level 3 neutralizers). Epitope mapping of plasma antibodies in some level 3 neutralizers has identified more than one neutralizing antibody specificity. A fundamental question is whether one or multiple specificities are responsible for the breadth of neutralization in broadly neutralizing plasma. Methods: We used near clonal memory B-cell cultures, EBV transformation and antigen-specific memory B-cell sorting to isolate two clonal lineages of bNabs from a single level 3 neutralizer (CH0219) infected with a subtype A virus. Results: Plasma from CH0219 neutralized 93% of clade A, B, C, AG, AE, G and D HIV-1 strains tested. Four VH3-20/Vk3-20 mAbs that were in the same clonal lineage (CH01-CH04) were isolated from CH0219 memory B-cells and neutralized 45% of HIV-1 strains. CH01-CH04 mAbs were neutralization sensitive to N160, F159, K169, K171 and I181 gp120 mutations, and crossblocked mAb PG9. Thus, CH01-04 mAbs were identified as V2/ V3 conformational antibodies that bound to an epitope similar to that of mAb PG9. Another clonal lineage (VH1-2/Vk1-33) of five antibodies (VRC-CH30-34) was isolated from CH0219 by resurfaced core 3 antigen-specific memory B-cell sorting. These antibodies were found to target the CD4-binding site and to neutralize 84% of HIV-1 strains. Two additional neutralizing mAbs were isolated from CH0219 against the V3 and gp41 regions that did not have significant breadth. Mixture of the CH01 and VRC-CH31 mAbs neutralized 93% of HIV-1 strains, including all those neutralized by the plasma sample. Conclusion: The CH0219 African donor made both V2/V3 conformational and CD4-binding site bNabs and the combination of these two specificities recapitulated the plasma neutralization breadth. This is the first documented case of two bNAbs targeting distinct HIV-1 envelope epitopes isolated from a single individual.

Background: VRC01-like antibodies achieve neutralization of over 90% of circulating HIV-1 strains by partial mimicry of CD4 in their heavy chain. While this mode allows for precise recognition by heavy chain of the initial site of CD4 attachment, the light chain cannot be fully accommodated and its recognition extends outside of the initial site of CD4 attachment into the variable regions including loop D and V5 that surround this site. Methods: How do VRC01-like antibodies accommodate variable regions on HIV-1 gp120 to achieve broad recognition and neutralization? To answer this question, we crystallized and studied the structural details of protein complexes between HIV-1 gp120 with diverse loop D and V5 regions and VRC01-like antibodies with different light chains. In particular, we analyzed VRC01-like antibodies with light chains from the kappa 3 family (VRC01 and VRC-PG04) as well as a VRC01-like antibody with a light chain from the kappa 1 family (VRC-CH31). Results: We found that the third complementarity determining regions (CDR) L3 of the light chains from both kappa 1 and kappa 3 families had very similar conformations, with a conserved glutamate and hydrophobic patch interacting with conserved regions of loop D and V5. To accommodate variable residues and glycosylation on these loops, the CDR L1 of kappa 3 origin used a loop shortened by a two-amino-acids deletion to avoid steric clashes. In contrast, CDR L1 of kappa 1 origin had no deletion in sequence, but adopted a conformation which circumvented potential clashes with loop D of gp120. Conclusion: In addition to using light chains of different family, VRC01-like antibodies utilize a conserved CDR L3 interaction as an anchor. The entire light chain pivots around the CDR L3 interaction, and does not recognize the loop D or V5 regions as much as avoid clashes, either through intrinsic flexibility or by affinity-maturation-evolved deletion.

POSTERS

116

AIDS Vaccine 2011

Topic 04: B Cell Immunology and Neutralizing Antibodies P04.41 LB


Crystal Structures of Human Anti-V2 mAbs 69730D and 8.9D and What We Can Learn From Their Antigen-Binding Sites
R. Pan5, J.M. Sampson5, B. Spurrier5, M. Totrov1, T. ONeal5, C. Williams5, S. Boliar2, S. Allen2, J. Mulenga3, J. Robinson4, C.A. Derdeyn2, M.K. Gorny5, S. Zolla-Pazner5, X. Kong5
1

Posters

P04.42 LB
454 Pryosequencing of B Cells From HIV-1 Infected Donors With Quaternary-StructurePreferring Antibodies
M. Pancera1, J. Zhu1, S. ODell1, X. Wu1, Y. Yang1, B. Zhang1, N. Doria-Rose2, M. Connors2, P. Moore3, S. Karim4, L. Morris3, M. Simek5, P. Poignard6, D.R. Burton6, W. Koff5, J.R. Mascola1, P.D. Kwong1
1 VRC/NIAID/NIH, Bethesda, MD, USA; 2NIAID/NIH, Bethesda, MD, USA; 3AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa; 4 CAPRISA, University of KwaZulu-Natal, Durban, South Africa; 5International AIDS Vaccine Initiative, New York, NY, USA; 6IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA

Molsoft LLC, La Jolla, CA, USA; 2Emory University, Atlanta, GA, USA; 3Zambia Emory HIV Research Project, Lusaka, Zambia; 4Tulane University Medical Center, New Orleans, LA, USA; 5NYU School of Medicine, New York, NY, USA
Background: The immunogenic V2 region of HIV-1 gp120 has been largely overlooked as a target for AIDS vaccine discovery. However, recent studies of sera from vaccinees in RV144 trial suggested that anti-V2 Abs were elicited and possibly contributed to protection. Structural understanding of human anti-V2 mAbs and their epitopes can facilitate design of immunogens. Methods: We determined crystal structures of Fab fragments of two human anti-V2 mAbs 697-30D and 8.9D, both at a resolution of 2.5 , and analyzed their antigen-binding sites (ABS) and possible modes of interactions with V1V2 of gp120. Results: MAb 697-30D, from a subtype B virus infected subject and encoded by VH1-69 gene, is a broadly cross-reactive anti-V2 mAb able to neutralize Tier 1 pseudoviruses; its epitope was mapped to conserved residues in V2. Structural analysis of Fab 697-30D revealed that its ABS consists of two distinct regions: (1) A surface pocket is located at the center of CDR loops formed by large aromatic residues, and it can accommodate residues with large side chains in the epitope identified by functional studies. (2) A convex hydrophobic surface is comprised of a cluster of CDR H2/H3 residues. Comparison with structures of other VH169 mAbs suggests that mAb 697-30D likely binds to the region of a short helix or a relatively flat surface of V2. Autologous neutralizing mAb 8.9D was isolated from a subtype C infected subject, and its epitope was mapped to the stem of V1V2. Its ABS is split by the upward positioned Tyr100b of CDR H3 into a positively-charged side and a negatively-charged side. Surface pockets in these regions can bind side chains of charged residues of V1V2, facilitating escape by mutations. Conclusion: Crystal structures of human anti-V2 mAbs provide structure-function insights of their epitopes. This information may contribute to rational design of immunogens targeting V2 region of gp120.

Background: Broadly neutralizing quaternary-structure-preferring antibodies such as PG9, PG16, and CH01-04 are found in selected HIV-1 infected donors. Some characteristics for these antibodies have been determined: they rarely bind monomeric gp120, they do not neutralize viruses grown in presence of kifunensine or with an N160K mutation. Sequences and structures of PG16 and other quaternary-structure-preferring antibodies indicated some similarities among all members of this class including long 3rd heavy chain complementary determining regions (CDR H3s) of over 24 residues, substantial affinity maturation (over 13% divergence in nucleotide sequence from germline), and VH3 gene family origin. Some members of the class, moreover, are tyrosine sulfated in their CDR H3 regions. Methods: Five million PBMCs (~500,000 B Cells) of HIV-1 infected donors that display serum reactivities consistent with the quaternary-structure-preferring phenotype were used to obtain cDNAs, which were PCR amplified with VH family primers and antibodies sequences determined by 454 pyrosequencing. Results: 200,000 to 1 million reads were analyzed based on sequences and structures signature for the quaternary-structurepreferring antibodies. Heavy chain sequences that had CDR H3s of 24 or more residues, displayed 13% or higher divergence from germline, and were predicted to be sulfated tyrosine were synthesized, paired with a light chain from a known quaternarystructure-preferring antibody, and the resulting reconstituted IgG tested for neutralization. Unfortunately, heavy/light chain chimeras between different quaternary-structure-preferring antibodies especially between antibodies from different donors did not display functional complementation, and we are currently overcoming this method by analyzing donors from which heavy/light antibody pairs have been previous identified. Conclusion: 454 pyrosequencing is a powerful tool that can be used to understand B cell development based on explicit lineages involving thousands of antibody sequences. While difficulties with functional complementation complicate the identification of novel quaternary-structure-preferring antibodies, the technology should allow maturation lineages of known antibody pairs to be deciphered.

AIDS Vaccine 2011

117

POSTERS

Posters
P05.01

Topic 05: B Cell Vaccine Concepts and Design P05.02


Neutralising Antibodies After Immunisation with the Transmembrane Envelope Proteins of HIV-1, HIV-2, Two Foamy Viruses, and Three Gammaretroviruses
J. Denner1, M. Mhle1, D. Kaulitz1, S. Langhammer1, U. Fiebig1, R. Behrendt1, A. Bleiholder2, M. Lchelt2, R. Kurth1, V. Morozov1
1

Enzyme Digests Eliminate Non-Functional Env from HIV-1 Particle Surfaces Leaving Native Env Trimers Intact and Viral Infectivity Unaffected
T. Tong1, E. Crooks1, K. Osawa1, J.M. Binley1
1

Torrey Pines Institute, San Diego, CA, USA

Background: HIV-1 viruses and virus-like particles (VLPs) bear aberrant Env species that could undermine the development of antibody responses against Env trimers, thereby blunting the ability of particles to elicit neutralizing antibodies. Here, we sought to better understand the nature of junk Env and to devise strategies for its removal. Methods: VLPs were analyzed by blue native PAGE-Western blot, SDS-PAGE-Western blot and in infectivity assays. Results: Native trimers are surprisingly stable in the face of harsh conditions, suggesting that junk Env does not arise from trimer dissociation. The limited gp120 shedding that can occur immediately after synthesis of primary HIV-1 Envs was found not to be caused by aberrant cleavage at the tandem gp120/ gp41 cleavage sites, which were cleave co-dependently. Our data suggest, that glycosylation may influence gp120 shedding. A major VLP contaminant of particles was found to consist of an early, monomeric form of gp160, termed gp160ER that bypasses normal protein maturation and trafficks directly into particles from the endoplasmic reticulum. gp160ER was found to bind two copies of mAb 2G12, consistent with its exclusively high mannose glycan profile. These findings prompted us to evaluate enzyme digests as a way to remove aberrant Env. Remarkably, sequential glycosidase-protease digests led to a complete or nearcomplete removal of junk Env from many viral strains, leaving trimers and viral infectivity largely intact. Conclusion: Trimer-VLPs may be useful neutralizing antibody vaccine immunogens.

Robert Koch Institute, Berlin, Germany; 2German Cancer Research Center, Heidelberg, Germany

Background: Broadly neutralising antibodies binding to the membrane proximal external region (MPER) of the transmembrane envelope (TM) protein gp41 of HIV-1 designated 2F5 and 4E10 have been isolated from HIV-infected individuals. All attempts to induce such antibodies failed until now. In contrast, in immunisation studies with the TM proteins of gammaretroviruses neutralising antibodies binding to the MPER of their TM protein were successfully induced. Methods: Recombinant proteins corresponding to the ectodomain of the TM proteins of HIV-1, HIV-2, the feline foamy virus (FFV), the primate foamy virus (PFV), and the gammaretroviruses feline leukaemia virus (FeLV), porcine endogenous retrovirus (PERV) and Koala retrovirus (KoRV) were used for immunisation of mice, rats and goats; binding and neutralising antibodies were determined. In the case of FeLV cats were immunised in addition and in vivo challenge experiments were performed. Results: Immunisation with the TM proteins of FeLV, KoRV and PERV resulted in neutralising antibodies binding to the MPER as well as to the fusion peptide proximal region of the corresponding TM protein. One epitope in the MPER showed a sequence homology with the epitope of 4E10 in gp41. The neutralising antibodies induced by the TM protein of FeLV protected cats from FeLV challenge. In contrast, immunisation with the TM proteins of HIV-1, HIV-2, FFV and PFV did not result in neutralising antibodies and no antibodies binding to the MPER were found. Conclusion: Whereas immunisation with the small TM proteins of different gammaretroviruses resulted in neutralising antibodies binding to the MPER, immunisation with the larger TM proteins of two lentiviruses and two foamy viruses failed. The reason for the difference is unclear and needs further investigation. Whereas the TM proteins of the gammaretroviruses are unglycosylated in the virus and those of the other viruses are glycosylated, all antigens used for immunisation were produced in bacteria and were non-glycosylated.

POSTERS 118 AIDS Vaccine 2011

Topic 05: B Cell Vaccine Concepts and Design P05.03


Immunogenicity of a Stable Clade C HIV-1 gp140 Envelope Trimer in Different Adjuvants in Guinea Pigs
J. Nkolola1, B. Chen2, M. Seaman1, S. Reed3, D. Carter3, D. Barouch1
1 2

Posters

P05.04
Defects In B Cell Intrinsic- and Extrinsic Factors Distinguish Novel 2009 H1N1 Vaccine Responders From Non-Responders Among HIV Infected Persons on ART
S. Pahwa1, S. Pallikkuth1, S.Y. Silva1, M. Fischl1, R. Pahwa1
1

Beth Israel Deaconess Medical Center, Boston, MA, USA; Childrens Hospital Boston, Boston, MA, USA; 3Infectious Disease Research Institute, Seattle, WA, USA

University of Miami Miller School of Medicine, Miami, FL, USA;

Background: The native envelope (Env) spike on the surface of human immunodeficiency virus type 1 (HIV-1) is trimeric, and thus trimeric Env vaccine immunogens are currently being explored in preclinical immunogenicity studies. We previously reported the binding and neutralizing antibody (NAb) responses in guinea pigs elicited by clade A (92UG037.8) and clade C (CZA97.012) Env gp140 trimer immunogens selected and engineered for optimal biochemical stability. Methods: To improve the NAb responses elicited by the clade C gp140 trimer in Ribi adjuvant, we assessed its stability and immunogenicity in four formulations of Glycopyranosyl Lipid Adjuvant (GLA); aqueous, liposome, emulsion and alum, as well as CpG, Emulsigen, and R848 adjuvants. Stability was assessed by size-exclusion chromatography upon trimer re-purification from each adjuvant. Immunogenicity was determined by ELISA and TZM.bl NAb assays after immunizing guinea pigs 3 times with 100 g of protein trimer in the various adjuvants. Results: By size-exclusion chromatography, the clade C gp140 trimer exhibited monodisperse peaks when re-purified from GLA formulations, CpG, Emulsigen, and R848 indicating conformational stability in these adjuvants. Ribi adjuvant, however, showed evidence of protein aggregation. Comparable binding antibody ELISA titers of 6.5 logs were observed in all groups after two immunizations. The four GLA formulations and combinations of CpG with either Emulsigen or R848 adjuvants, all elicited potent, cross-clade NAb responses against select tier 1 viruses that were comparable to Ribi adjuvant. Against the more stringent tier 2 clade C viruses, the groups that received the GLA emulsion and aqueous adjuvants generated limited but clearly detectable tier 2 responses as compared with Ribi adjuvant. Conclusion: These data demonstrate the immunogenicity of our stable, clade C gp140 trimer in different adjuvant systems. The effects of adjuvants on conformational structure and integrity may be important in the selection of adjuvants for optimal induction of Env specific NAbs.

Background: This study investigated innate and adaptive immune responses associated with antibody (Ab) responsiveness to a single dose of non-adjuvanted H1N1/09 vaccine in 17 virologically suppressed HIV infected patients and 8 healthy controls (HC). H1N1 Ab titers of >1:40 units/4 fold rise developed in all HC but in only 9/17 patients. Methods: PBMC and serum were isolated from venous peripheral blood pre-vaccination (T0) and post vaccination on day7 (T1) and day28 (T2). Immunologic assessments were made in fresh PBMC by multiparameter flow cytometry and in cryopreserved PBMC/ serum by ELISPOT/ ELISA. Results: At T1, HC and vaccine responder (R) patients developed approximately 3 fold expansion of plasmablasts and >4 fold increase in spontaneous H1N1 antibody secreting cells (ASC). At T2, HC and R patients had expansion of memory B cells (HC, 1.4 fold; R, 2.5 fold) with increases in ex-vivo H1N1-stimulated IgG ASC, innate immune factors BAFF (B cells activating factor), APRIL (a proliferation-inducing ligand) and T cell cytokine interleukin (IL)-21. Concurrently IL-21R and TACI (transmembrane activator and calcium-modulator and cyclophilin ligand interactor) were upregulated and BAFFR downregulated on B cells. Frequency of CD4+CXCR5+ T follicular helper like cells increased (HC 1.3 fold, R 1.4 fold). Vaccine non-responder (NR) patients did not manifest these immunologic responses. At T0, R and NR patients were equivalent in mean age, plasma HIV RNA, CD4 and CD8 T cell counts and CD20+ B cells. Phenotypic memory B cells were equivalent (but lower than HC) between R and NR patients but frequency of BAFFR+ and TACI+ B cells were higher in R patients. Conclusion: Failure to mount H1N1 Ab responses was associated with multiple deficits of innate and adaptive immunity in association with altered B cell phenotype in otherwise stable HIV infected patients. These findings point to new avenues for investigation in future vaccine strategies.

AIDS Vaccine 2011

119

POSTERS

Posters
P05.05

Topic 05: B Cell Vaccine Concepts and Design P05.06


PG16 Binding Early Transmitted HIV-1 Clade C Virus 16055 Soluble Envelope Glycoprotein as Candidate Vaccine Immunogen
D. Wagner5, J. Gaekwad1, S. Narpala5, A. Carpov5, M. Pancera2, L.M. Walker3, S. Hoffenberg5, P.D. Kwong4, J.R. Mascola4, D.R. Burton3, W.C. Koff5, R. King5, R.T. Wyatt1, S.K. Phogat5
1

Predicted Antigenic Map of the V2 Loop


E. Shmelkov2, R. Abagyan1, J. Swetnam2, S. Zolla-Pazner2, T. Cardozo2
1 2

University of California, San Diego, San Diego, CA, USA; New York University School of Medicine, New York, NY, USA

Background: The V2 loop of HIV-1 strains is immunogenic and exhibits an 47 receptor binding site. Methods: We inferred an antigenic map of the V2 loop by controlling for its length distribution across circulating strains 1) by multiply aligning only the sequences of the most common V2 loop length (40 amino acids) and 2) by taking length into account in deriving a consensus sequence. Ab initio folding was used to evaluate the local secondary structure preferences of a middle conserved block of amino acids spanning the 47 receptor-binding site. The sequence conservation pattern, known glycosylation sites and known anti-V2 Ab binding sites were then mapped. Results: The V2 loop exhibits an asymmetric distribution of chain lengths with few or no chains less than 38 amino acids and most chains being 40 amino acids in length or longer. There are three blocks of conserved amino acids in the V2 loop, and most of the length variation occurs between the second (containing the 47 receptor binding motif) and third block. Ab initio folding demonstrated that the second block has a preference for alpha-helical secondary structure. Mapping the amino acid conservation pattern of the second conserved block onto an alpha-helix clusters several conserved V2 amino acids into a single 3D region coinciding with previously published mutations that affect the binding of the anti-V2 Ab 697-D and coinciding with the 47 receptor binding motif. Glycan chains or residue positions with poor amino acid conservation among circulating strains are predicted to obscure most of the remainder of the molecular surface. Conclusion: A 3D map of the second sequence conserved block of the V2 loop is an antigenic map exhibiting an exposed site composed of amino acids forming the a4b7 receptor binding site that are also part of the epitope of a known human antiHIV antibody.

IAVI NAC Center at TSRI, La Jolla, CA, USA; 2Vaccine Reserach Center, NIAID, NIH, Bethesda, MD, USA; 3The Scripps Reserach Institute, LaJolla, CA, USA; 4Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA; 5International AIDS Vaccine Initiative, Brooklyn, NY, USA
Background: HIV-1 native envelope glycoprotein (Env) spike is the principal determinant for neutralizing antibodies. Antibodies PG9, PG16 and CH01-05 are broadly neutralizing antibodies (bnAbs), which bind an epitope preferentially expressed on trimeric Env. The epitope appears to span conserved regions of the major variable loops 2 and 3 present on the gp120 subunit, with asparagine at position 160 being a critical determinant. These bnAbs can help identification of soluble Env forms that mimic Env trimer to which these antibodies bind preferentially. Methods: Shed-gp120 and bnAbs (PG9, 16 and b12) binding was used for identification of soluble Env forms displaying relevant conformation/structure similar to that presented on the native spike. The selected gp120 were characterized by biochemical and biophysical methods. Results: We have identified HIV clade C early transmitted virus 16055 Env that binds with nanomolar affinity to PG9, VRC01 and CD4IgG. 16055 Env when converted to gp140 trimers binds trimer-specific PG9, PG16, CD4 binding site-specific VRC01, PGV04, b12 and gp41-specific 4E10 bnAbs. High affinity binding of 16055 gp140 to trimer specific PG16 antibody suggests that these molecules present conformation of the variable loops 2 and 3 representative of functional HIV-1 Env. Importantly, 16055 virus shows binding and neutralization by germ line precursor mimic for PG9. Mutagenesis of asparagine (N) at position 160 and glutamic acid (E) at position 168 abrogates PG9 and PG16 binding on the 16055 Env but do not effect CD4IgG binding. Furthermore, the PG9 and PG16 binding and neutralization property of 16055 was transferred to 16936 virus by V1V2 and V3 loop swap. 16936 virus, in the absence of loop swap, is resistant to PG9 and PG16. Conclusion: 16055 soluble Env proteins mimic some conformational characteristics of the native spike as indicated by PG16 antibody binding, 16055 Env will help understand the V1V2V3 loops conformation and as novel HIV immunogen.

POSTERS 120 AIDS Vaccine 2011

Topic 05: B Cell Vaccine Concepts and Design P05.07


Native HIV-1 Sequential Envelope Quasispecies as a Novel B Cell Vaccine Concept
D.C. Malherbe1, N. Doria-Rose2, L. Misher2, T. Beckett2, W. Blay-Puryear2, J. Schuman3, Z. Kraft2, J. OMalley1, M. Mori1, I. Srivastava4, S. Barnett4, S. Kalams5, L. Stamatatos2, N.L. Haigwood1 Oregon Health and Science University, Beaverton, OR, USA; 2Seattle BioMed, Seattle, WA, USA; 3GE Healthcare, Piscataway, NJ, USA; 4Novartis, Cambridge, MA, USA; 5 Vanderbilt University, Nashville, TN, USA
1

Posters

P05.08
Robust Multi-Clade Cellular and Humoral Immune Responses in Rhesus Macaques Following Optimized Consensus DNA Vaccination
N. Hutnick1, D.F. Myles1, J. Yan2, A.S. Khan2, K. Muthumani1, N.Y. Sardesai2, D.B. Weiner1
1

University of Pennsylvania, Philadelphia, PA, USA; 2Inovio Pharmaceuticals, Inc., Blue Bell, PA, USA

Background: HIV-1 evolves rapidly within the host, resulting in the development of diverse HIV-1 variants called a viral quasispecies population. Envelope (Env) is the only target of neutralizing antibodies (NAbs), which can prevent infection of target cells. NAbs increase in titer and affinity over time as Env diverges. A major goal of HIV-1 vaccine efforts, so far elusive is the design of Env-based immunogens effective at eliciting broad NAbs (BNAbs). We hypothesize that B cells are programmed to develop BNAbs by exposure to Envs presented by the viral quasispecies variants. Methods: We cloned 15 env genes from different timepoints in a macaque that developed BNAbs following SHIV-SF162P4 infection. Rabbits were immunized (gp160-DNA prime, gp140protein boost) with: (1) sequential env clones (Sequential); (2) a cocktail of env clones (Mixture); or (3) a single env variant (Clonal). We identified HIV+ subjects with BNAbs and developed DNA and protein vaccines based on these quasispecies. Results: All groups immunized with the SHIV envs generated strong ANAbs and modest HNAbs against Tier-1 viruses. Late autologous clones were more difficult to neutralize than early ones. Specific PNGs in V1, V2 and V4 are involved in neutralization resistance, as are key amino acid residues. The Mixture and Sequential strategies replicated the epitope targeting of the V3 loop seen in the macaque. The Sequential and the Clonal strategies elicited higher antibody affinity maturation than the Mixture. Vaccines based on the human HIV Env quasispecies are in testing. Conclusion: Taken together, these results suggest that the Sequential modality elicits responses that are more similar to those developed by subjects with breadth. This study is the first to explore the use of multiple native HIV-1 Env variants as immunogens and to show it is possible to educate the immune system by exposing it to a native HIV-1 quasispecies derived from an individual with BNAbs.

Background: Advancements to the DNA vaccine platform have improved the magnitude and quality of the immune response achieved in both small and large animal models. Data presented at the recent Keystone HIV Vaccine meeting by the HVTN from our HVTN 080 trial demonstrates that the CD4 and CD8 cellular responses induced by the combination of a highly optimized, cytokine adjuvanted, HIV-1 DNA vaccine (PENNVAX) delivered by EP (E-DNA) in humans at are as good or better than those induced by viral vector systems alone, or in prime boost combinations. Recently, we have observed that E- DNA outperforms adjuvanted envelope protein for induction of neutralization antibody titers, results improved with a DNA prime protein boost. Here we sought to define the potential of a DNA prime for inducing both cellular and humoral responses in non-human primates prior to a protein boost. Methods: Three groups of five rhesus macaques received 1.0 mg consensus pVax HIV-1 M gag, HIV-1 M pol, and HIV-1 B either IM 0.5 Amps, ID 0.2 Amps, or ID 0.1 Amps. A Fourth group of five macaques included HIV-1 Env A, C, D, and A/E IM. DNA was delivered at weeks 0, 6, 12 and 18. Results: CD8+ responses greater than 1% were observed following only two doses of E-DNA in all groups. Multi-envelope vaccination induced multiclade T-cell responses as well a tenfold increase in antibody binding titers to HIV-1 B env compared with single clade groups. Antibody titers greater than 1:1000 were consistently achieved with E-DNA alone. Conclusion: These data demonstrate that optimized E-DNA can induce robust immune responses in the NHP model. The inclusion of multiple envelope vectors induces cross-reactive responses that increase the breadth and magnitude of vaccine induced responses. Additionally, E-DNA priming can induce potent antibody response that may be boosted by a recombinant protein boost.

AIDS Vaccine 2011

121

POSTERS

Posters
P06.01

Topic 06: Clinical Trial Site Challenges P06.02


Barriers of Enrollment in HIV Vaccine Trials: A Review of HIV Vaccine Preparedness Studies
S. Dhalla1, G. Poole1
1

Motivators of Enrollment in HIV Vaccine Trials: A Review of HIV Vaccine Preparedness Studies
S. Dhalla1, G. Poole1
1

University of British Columbia, Vancouver, Canada

University of British Columbia, Vancouver, Canada

Background: HIV vaccine preparedness studies (VPS) are important precursors to HIV vaccine trials. As well, they contribute to an understanding of motivators and barriers for participation in hypothetical HIV vaccine trials. Motivators can take the form of altruism and a desire for social benefits. Perceived personal benefits, including psychological, personal, and financial wellbeing, may also motivate participation. Methods: We performed a systematic review of HIV VPS using the Cochrane Database for Systematic Reviews, Medline/Pubmed, Embase, and Google Scholar. Two people independently searched the literature for individual HIV VPS that examined motivators of participation in a hypothetical HIV vaccine trial. The denominators employed in the literature varied across studies, and these were standardized to the number of respondents per survey item, regardless of their willingness to participate (WTP) in an HIV vaccine trial. The Organization for Economic Cooperation and Development (OECD) countries and the non-OECD countries were compared with respect to these motivators. Results: We retrieved eight studies on social benefits (i.e., altruism) and 11 studies on personal benefits conducted in the OECD countries, as well as 19 studies on social benefits and 20 studies on personal benefits in the non-OECD countries. Various different forms of altruism were found to be the major motivators for participation in a hypothetical HIV vaccine trial in both the OECD and the non-OECD countries. In a large number of studies, protection from HIV was cited as a personal motivator for participation in a hypothetical HIV vaccine trial in the OECD and the non-OECD countries. Conclusion: This is the first comprehensive review examining motivators of participation for an HIV vaccine trial. Knowledge of motivators can inform and target recruitment for HIV vaccine trials, though it must be remembered that hypothetical motivators may not always translate into motivators in an actual vaccine trial.

Background: Barriers to participation in an HIV vaccine trial have been examined in many HIV vaccine preparedness studies (VPS). These barriers can be understood in terms of the locus of the barrier (personal vs. social) and the nature of the barrier (risk vs. cost). Another type of barrier is perceived misconceptions. Methods: We performed a systematic review of HIV VPS using the Cochrane Database for Systematic Reviews, Medline/ Pubmed, Embase, and Google Scholar. Two people independently searched the literature for HIV VPS that examined barriers of participation in a hypothetical HIV vaccine trial, using the same search strategy. We categorize these barriers, and compare barriers between the Organization for Economic Co-operation and Development (OECD) countries and the non-OECD countries. Risk was operationalized in terms of possible outcomes; costs in terms of very probably outcomes. Results: In the OECD countries, we retrieved 18 studies reporting personal risks (PR), 7 studies reporting social risks (SR), 6 studies reporting personal costs (PC), and 15 studies reporting misconceptions. In the non-OECD countries, we retrieved 22 studies reporting PR, 16 studies reporting SR, 14 studies reporting PC, 1 study reporting social costs (SC), and 19 studies reporting misconceptions. Important PR were adverse effects and vaccine-induced seropositivity, and temptation to have unsafe sex in men who have sex with men (MSM). Discrimination was a common SR. Fear of needles and time commitment were important PC, and family commitments were a SC in one non-OECD country. Distrust of institutions and HIV infection from the vaccine were common misconceptions. Conclusion: This is a comprehensive review to participation in an HIV vaccine trial. Both the OECD and non-OECD countries have similar barriers, and peoples decisions to participate in a clinical trial involve multiple barriers. However, these barriers apply to hypothetical HIV vaccine trials, and barriers for actual vaccine trials need more assessment.

POSTERS 122 AIDS Vaccine 2011

Topic 06: Clinical Trial Site Challenges P06.03


Development of CAVD/EQAPOL Long-Term PBMC Repository (07-10) @ DUMC; T-Cell Functionality as Measured by ELISpot and Flow Cytometry Is Maintained Ov
G. Ferrari1, J. Cox2, A. Garcia3, W. Rountree3, C. DeCastro4, R. Louzao3, B. Liebl3, A. Sambor5, A. Sanchez3, M. Sarzotti-Kelsoe6, J. Lane7, P. DSouza8, R. Koup9, T. Denny10 Duke University, Durham, NC, USA; 2International AIDS Vaccine Initiative, Rockville, MD, USA; 3Duke Human Vaccine Institute, Duke University, Durham, NC, USA; 4Duke Institute for Genome Sciences and Policy, Duke University, Durham, NC, USA; 5Foundation for the National Institutes of Health, Bethesda, MD, USA; 6Center for AIDS Research Central QA Unit, Duke University, Durham, NC, USA; 7Vaccine Clinical Research, Division of AIDS, National Institute of All, Bethesda, MD, USA; 8Vaccine Clinical Research, Division of AIDS, National Institute of AIl, Bethesda, MD, USA; 9Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA; 10Duke Human Vaccine Institute, Center for HIV/AIDS Vaccine Immunology, Durham, NC, USA
1

Posters
P06.04
Prevalence, Incidence, Risk Factors and Willingness to Participate in HIV Vaccine Trials Among Gay and Bisexual Men and Transgender Persons Seeking HI
S. Chariyalertsak1, C. Beyrer2, N. Kosachunhanan3, P. Saokhieo3, R. Songsupa3, A. Wongthanee3, C. Chariyalertsak4, S. Visarutratana4
1

Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand; 2Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; 3RIHES, Chiang Mai University, Chiang Mai, Thailand; 4Ministry of Public Health, Chiang Mai Provincial Health Office, Chiang Mai, Thailand

Background: HIV prevalence among men who have sex with men (MSM) and transgender (TG) persons is high and increasing in Chiang Mai, northern Thailand. Methods: In 2008-09, gay and bisexual men and transgender women attending HIV testing and counseling at an MSM friendly clinic in Chiang Mai (PIMAN Clinic) were interviewed and tested for HIV and STI. We sought to investigate demographic, socioeconomic, sexual behavior and interest in future HIV prevention trials among these participants and to conduct prescreening for the iPrEx pre-exposure chemoprophylaxis trail. Univariate and multivariate regression analyses were done to assess associations with HIV infection. Results: A total of 551 MSM clients (56.1% gay, 25.4% TG, and 18.5% bisexual (BS)) were enrolled. The mean age was 23.9 years. HIV prevalence among MSM overall was 12.9% (71/551; 16.5% among gay men, 9.3% among TG, and 6.9% among BS. Consistent use of condom was low, 33.3% at last insertive anal sex and 31.9% in last receptive anal sex. Most, 55.4%, had prior HIV testing, and 64.6% had VCT within 1 year. Sex role segregation was marked, with nearly all TG reporting only receptive anal sex; nearly all BS reporting exclusive insertive sex, and most gay men reporting both roles. Interest in participation was high at 69.7%, for HIV vaccine trials. Interest in trial participation was not associated with HIV infection or risks. HIV was independently associated with being gay identified, aOR 2.8, p= 0.037; and with being aged 25-29, aOR 2.7, p= 0.027. Among repeat testers, HIV incidence was 8.2/100 PY, 95% CI, 3.7/100PY to 18.3/100PY. Conclusion: These young and at risk populations are in urgent need of novel HIV prevention strategies and are willing to participate in HIV vaccine research.

Background: Testing of clinical samples to evaluate vaccineinduced responses by endpoint laboratories requires extensive standardization and validation of the assay and corresponding controls to monitor assay performance. Samples with wellcharacterized functionality are needed for assay development and comparison to preexisting assays. To provide these samples, we have established a PBMC repository in which cells are processed and stored to retain viability, recovery and functionality. The repository includes HIV seropositive and seronegative donor samples with a wide range of CEF-, CMV-, and HIV-specific functional responses. Methods: PBMCs were collected by leukopheresis from 223 donors, isolated by density gradient separation, and cryopreserved using a rate-controlled freezer. For all samples, viability, recovery, and functionality were assessed by IFN- ELISpot withinin 4 weeks of freezing. We evaluated viability, recovery, and functionality every 6 months in 5 samples with different levels of responses and monthly in 1 low/medium responder. Finally, in a subset of 70 samples functionality was measured by both ELISpot and IFN-/IL-2 ICS assays. Results: Viability of 97.7% of the samples was above 80%, and the recovery of 90.5% was above 66% after thawing and resting the cells overnight. For the 5 low/moderate and high CEF and CMV responders, there was no specific trend suggesting a loss in viability or recovery over time. Functionality was retained in 4 of the 5 samples. No specific trends suggesting degradation for any parameter were observed for the low/moderate responder measured monthly. A significant correlation was observed for CEF (R2=0.91, p < 0.0001) and CMV (R2=0.93, p < 0.0001) IFN- responses measured by ELISpot and ICS. Since 2007, approximately 4,300 PBMC vials have been distributed to 23 laboratories, worldwide, to perform assay development, validation and quality assurance. Conclusion: In conclusion, we have developed PBMC repository for use by endpoint laboratories for assay validation and development across different networks.

AIDS Vaccine 2011

123

POSTERS

Posters
P06.05

Topic 06: Clinical Trial Site Challenges P06.06


Factors Influencing Recruitment, Retention and Seroconversion Rates in MSM at High Risk for HIV Infection in Lima and Barcelona
P. Coll1, S. Perez-Alvarez1, A. Tataje2, C. Ganoza2, A. Leon3, F. Pujol4, M. Meulbroek4, I. Bravo5, E. Fernndez3, J. Casabona6, J. Gatell3, B. Clotet1, J. Snchez2, C. Brander7 Institut de Recerca de la Sida-HIVACAT, Barcelona, Spain; Asociacion Civil Impacta Salud y Educacion, Lima, Peru; 3 Hospital Clinic, Barcelona, Spain; 4Hispanosida, Barcelona, Spain; 5Fundaci Lluita contra la SIDA, Barcelona, Spain; 6 CEEISCAT, Barcelona, Spain; 7Instituci Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
1 2

Challenges in Consenting Sex Workers, Transgenders and Men Who Have Sex with Men into an HIV Acute Infection Study in Pattaya, Thailand
P. Charuthamrong , W. Kaeratiswetanun , T. Yamkram , M.W. Benenson1, P.A. Morgan1, S. Nitayaphan1, C. Eamsila1, V. Tungsakul1, S. Sriplienchan1, M. Robb2, N. Thaitawat1
1 1 1

Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 2US Military HIV Research Program, Rockville, MD, USA
1

Background: The acute stages of HIV-1 infection influence disease progression. Understanding the earliest host-virus interactions is criticial to vaccine development. The Armed Forces Research Institute of Medical Sciences is conducting an acute infection study in Pattaya. Starting mid-2009, it recruited among sex workers, transgenders, and men who have sex with men. In addition to the main consent form, there are five other forms for specific procedures and withdrawal. Several factors can influence volunteers understanding of the trial and decision to participate - education, alertness, peer pressure, expectation of benefits. Specific measures are implemented to address these factors. Methods: NGOs help craft the language in the consent forms, review recruitment materials and design community mobilization activities. They informally pre-screen potential volunteers for understanding and willingness during outreach. At the first visit, pre-screening for alertness and reasons for enrolling is conducted. If volunteers are not ready, the consent form is given to review and an appointment is made to return. For those determined ready, a one-on-one review of the main consent followed by a test of understanding is done. Once enrolled, counseling sessions, held quarterly, are used to review key points of the trial and discuss factors affecting their participation. Results: 910 people were seen, of whom 455 were screened. All passed the test of understanding. 322 were enrolled and could state the trials objective and rationale for the research methodology during counseling sessions. Primary reasons for participation reported at screening are want to know HIV status 201 respondents and want to do good 82. Most volunteers reported the main consent was long and technical. Conclusion: Careful screening and an ongoing process that underlines the trials objective and research methodology can help volunteers maintain understanding of their participation. However, common factors related to sex work affect level of understanding and rationale for participation.

Background: The availability of well-defined cohorts with high retention rates is crucial for HIV vaccine development. Aside from representing suitable cohorts for vaccine efficacy trials, closely monitored cohorts of individuals at elevated risk for HIV infection also offer access to individuals with acute HIV infection and highly exposed persistently seronegative subjects (HEPS), for the study of factors associated with increased or reduced susceptibility to HIV acquisition. Methods: Men who have sex with men (MSM) were recruited in Lima and Barcelona by advertising in community-based HIV detection centres, MSM venues and magazines. Individuals at elevated risk were selected through an initial risk-assessment questionnaire. Participants were followed on a quarterly basis for HIV infection. In the Barcelona cohort, participants were also screened for the main sexually transmitted infections. Data from questionnaires and blood samples were collected at each visit. Results: 237 subjects were enrolled in Lima. The loss of followup was 20% in the first year (the majority lost within the first 6 months) and then stabilized for year 2 (10%) and 3 (12%). HIV incidence was 4%, with most of the new infections (87%) occurring between visits 2 and 4, resulting in a seroconversion rate of 9% in the first year. In the Barcelona cohort, 160 individuals were recruited thus far, with 3 seroconversions observed to date (all occurring within the first three visits). Retention rate was 92%. Conclusion: Prospective tailoring of cohort-specific retention strategies in consultation with community members and applying initial questionnaires to assess high risk helps to establish cohorts with elevated seroconversion rates. The concentration of new infections at earlier time points may facilitate cohort management (in terms of duration, number of enrolees) to capture new infections and, importantly, may reflect an educational effect among the enrolees during their participation in these studies.

POSTERS 124 AIDS Vaccine 2011

Topic 06: Clinical Trial Site Challenges P06.07


Providing Care and Education Prior to Engaging in an HIV Vaccine Trial, Bamako Mali, West Africa
L. Levitz5, K. Sangare1, K. Tounkara2, F. Siby Diallo3, S. Traore2, Y. Kone2, O. Koita1, J. Rozehnal4, Z. Koty2, M. Rochas2, A. De Groot5
1

Posters
P06.08
Education and Research Initiatives for the Mobilization of African-American Faith Communities for HIV/AIDS Prevention and HIV Vaccine Awareness
M.C. Keefer1, C.A. Bunce1, S.J. Thompson2, S.F. Wakefield3, R.C. Sanders II4 University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; 2Metropolitan Interdenominational Church Technical Assistance Network, Nashville, TN, USA; 3 HIV Vaccine Trials Network Legacy Project, Seattle, WA, USA; 4Metropolitan Interdenominational Church Technical Assistance Network, Nashville, TN, USA
1

LBMA-Laboratoire de Biologie Moleculaire Appliquee, Bamako, Mali; 2GAIA Vaccine Foundation, Bamako, Mali; 3DRSDirection Regionale de la Sante, Bamako, Mali; 4Institute for Immunology and Informatics - University of Rhode Island, Providence, RI, USA; 5EpiVax Inc., GAIA Vaccine Foundation, URI, Brown Medical Department, Providence, RI, USA

Background: The GAIA Vaccine Foundation (VF) has been collaborating with Malian HIV clinicians, scientists and community-based organizations to prepare a site for Phase I-III HIV vaccine trial in Sikoro, a multi-ethnic neighborhood of Bamako, Mali where more than 40,000 people live; 90% are illiterate and roughly 50% are unemployed. Methods: GAIA VF has worked to improve HIV prevention through MTCTP, access to HIV treatment and peer education. HIV specialists working at the Hope Center Clinic (HCC), a communitybased infirmary located in Sikoro, have improved prevention and set up a comprehensive clinical and biological chart review of patients treated with ARV. The HCC was built in 2007-08 and became the first village-based clinic with an HIV care program in February 2009. Results: 8,210 pregnant women at the village clinic have been tested for HIV; 174 were diagnosed with HIV. MTCTP was offered to all women; 99% accepted. Until 2009, HIV+ patients at the clinic were transported to off site for treatment. >120 patients have enrolled since 2009 at the new on-site care center, of which 59 are on treatment. GAIA VF has also performed three KAP studies in Bamako, in collaboration with the DRS, to evaluate baseline levels of HIV knowledge and willingness to participate in an HIV vaccination trial. Of 399 persons surveyed in Sikoro, 78% were willing to participate (WTP) in an HIV vaccine trial, 65% were WTP in a Malaria vaccine trial and 61% were WTP in a TB vaccine trial. Women were more WTP (81%) than men (76%). Conclusion: Providing HIV care and education allowed GAIA VF to reinforce the rapport between clinic staff and community members, assess HIV knowledge and misconceptions and WTP in a vaccine trial. This groundwork will help GAIA VF set up a Phase I-III HIV vaccine trial site in this region of Mali, West Africa.

Background: The African-American (AA) community bears a disproportionate burden of HIV in the United States and the AA church must assume a leadership role in stemming the epidemic. However, AA clergy may feel ill-equipped to engage the issue without HIV education that aligns with their faith tradition. We established a collaborative program to provide AA clergy with the understanding needed to lead discussions on HIV-related issues, including vaccine research, with their congregations. Methods: Capacity building for AA church leadership in Rochester, NY was provided through a series of seminars/group discussions between local clergy and a CDC-supported spiritual leader, Rev. Edwin Sanders II, from MICTAN in Nashville, TN. Additional support was provided by the NIAID-funded HVTN Legacy Project, the UR HVTN site and 2 local communitybased organizations with faith initiatives. Clergy participated in four monthly seminars presented by nationally-recognized AA leaders in the church/academia who addressed barriers to clergy engaging in HIV ministry. Participants were required to attend at least 3 seminars to be eligible to attend the annual MICTAN Faith Academy meeting in Nashville; those who completed written assignments received certification from MICTAN. Results: The program was well-received by Rochesters AA clergy. Fifty-one clergy attended the introductory seminar, 20 fulfilled requirements to attend the Faith Academy, 19 travelled to Nashville and 12 received certification. Pastors embraced universal core values for engaging in inclusive HIV/AIDS ministries and expressed eagerness to continue collaborating with UR and MICTAN. Conclusion: Many AA clergy in Rochester, NY were willing to address HIV/AIDS issues when provided spiritually-grounded HIV/AIDS training. The monthly series created a safe environment for ongoing dialogue and an opportunity for relationships to develop. The programs success hinged on sustained collaborative efforts between CDC, NIH, state and locally-funded partners to build the trust required for AA clergy to become invested in HIV vaccine research.

AIDS Vaccine 2011

125

POSTERS

Posters
P06.09

Topic 06: Clinical Trial Site Challenges P06.10


Incidence of HIV Among Youths in Maputo City, Mozambique: A Cohort Study
2

Reference Values for Clinical Laboratory Parameters Among Youths in Maputo City, Mozambique
N.R. Tembe , E.E. Viegas , N. Sitoe , E. Alfai , E. Mucavela , N. Osman2, S. Andersson3, I.I. Jani1, C. Nilsson4
1 1 1 1

E.N. Viegas1, N. Tembe1, E. Gonalves2, E. Macovela2, N. Ismael1, C. De Schacht1, N. Bhatt1, C. Nilsson3, G. Biberfeld3, S. Andersson4, N. Osman2, I.V. Jani1
1 Instituto Nacional de Sade de Moambique, Maputo, Mozambique; 2Maputo Central Hospital, Maputo, Mozambique; 3Swedish Institute for Communicable Disease Control and Karolinska Inst, Stockholm, Sweden; 4 Department of Laboratory Medicine, rebro University Hospital, rebro, Sweden

Instituto Nacional de Sade de Moambique, Maputo, Mozambique; 2Hospital Central de Maputo, Maputo, Mozambique; 3Department of Laboratory Medicine, rebro University Hospital, rebro, Sweden; 4Swedish Institute for Communicable Disease Control and Karolinska Institute, Stockholm, Sweden
1

Background: Common practice in Mozambique is to use reference values from Europe or US provided by clinical laboratory assay systems. A planned phase I/II HIV vaccine trial in youths necessitated the establishment of reference values for clinical laboratory parameters. Methods: A cross sectional study was conducted at a youth clinic in Maputo City, Mozambique. A total of 196 participants (1824 years old) were enrolled, of which 144 were female and 52 male. Medical staff collected clinical histories using standardized forms. Immunophenotyping was performed using a singleplatform lyse-no-wash procedure with Trucount tubes and MultiTest (CD3FITC/CD8PE/CD45perCP/CD4APC) reagents on a FACSCalibur cytometer (all from Becton-Dickinson). A complete blood count and differential was performed using a Sysmex KX-21N Hematology Analyzer. Serum chemistry was performed using a Vitalab Selectra Junior (Vital Scientific). Median and 2.5th-97.5th percentile reference ranges were established for immunology, hematology and chemistry values. Results: Median levels were as follows: hemoglobin, 13.75 (male) and 11.2 (female) g/dL (p=0.0001); platelets, 220x103/L (male) and 264x103/L (female) (p=0.0028); leukocyte (WBC) counts, 4.9x103/L (male) and 5.45x103/L (female) (p=0.018); absolute lymphocyte counts, 1869 cells/L (male) and 2070 cells/L (female) (p=0.018); absolute CD4 T cell count, 698 cells/L (male) and 828 cells/L (female) (p=0.0001); absolute CD8 T cell count, 467 cells/L (male) and 474 cells/L (female) (p=0.8821); bilirubin, 0.65 (male) and 0.26 (female) mg/dl (p=0.0001); albumin, 5.02 (male) and 4.78 (female) g/L (p=0.0001); creatinine, 0.99 (male) and 0.74 (female) mg/dl (p=0.0127); aspartate aminotransferase (AST), 25.05 (male) and 20.60 (female) U/L (p=0.0001); and, alanine aminotransferase (ALT), 15.40 (male) and 11.00 (female) U/L (p=0.0001). Conclusion: We have reported reference values for hematology, immunology and biochemistry parameters in youths (18-24 years) which will inform inclusion criteria and evaluation of adverse events in planned HIV vaccine trials in Maputo, Mozambique.

Background: HIV prevalence in 15-49 year old individuals in Mozambique is 11.5%, being 16.8% in Maputo City. Across the country, HIV prevalence is higher in women than in men (13.1% vs 9.2%), peaking at ages 25-29 and 35-39 in women and men, respectively. This study aimed at determining the incidence of HIV and prevalence of other sexually transmitted infections in young adults, and at creating a cohort of youths for future HIV vaccine trials in Maputo City. Methods: Young adults from both genders aged 18-24 (n=1380) were recruited at a youth clinic and followed-up for 12 months. Clinical, demographic and behavioural data were collected using standardised questionnaires. HIV testing was conducted at screening and every four months using rapid antibody and PCR assays. Serological testing for hepatitis B (HBV) and syphilis was performed at screening. Viral load was measured in acute HIV cases using real-time PCR. Results: Volunteers had a median age of 20.9 years and were mostly female (77.3%). All volunteers had some formal education, with 98.6% having at least the secondary level. Almost all (98.8%) of study participants reported at least one previous sexual intercourse, with a median age of 17 years at the first intercourse. At screening, the prevalence of HIV, HBV and syphilis was 5.1% (95%CI 3.97%-6.31%), 11.1% (95%CI 9.31%-12.91%) and 0.51% (95%CI 0.13%-0.89%), respectively. The prevalence of HIV-HBV and HIV-syphilis co-infections was 0.59% (95%CI 0.16%-1.04%) and 100%, respectively. In youths that tested HIV-negative at screening (n=1309), the HIV incidence was 1.14/100 person-years (95%CI 0.57-2.27). All acute HIV infections (n=8) occurred in female individuals. HIV plasma viral load in these patients ranged from 9370-188,419 copies/mL. Conclusion: HIV incidence in this cohort of youths in Maputo City is relatively low. This cohort is suitable for the conduct of phase I/II vaccine trials.

POSTERS 126 AIDS Vaccine 2011

Topic 06: Clinical Trial Site Challenges P06.11


The Impact of a Multi-Faceted Training and Mentoring Program in HIV Risk Reduction Counseling at Global HIV Vaccine Trial Sites
D. Flood1, J. Fuchs2, E. Dossett3, E. Wolf4, B. Metch5
1

Posters
P06.12
Hepatitis B Virus (HBV) Immunity After Vaccination in a Preparatory Cohort Study Among Men Who Have Sex with Men, Bangkok, Thailand, 20062009
T.H. Holtz1, B. Raengsakulrach1, W. Chonwattana1, W. Thienkrua1, J. McNicholl2, W. Wimonsate1, S. Chaikummao1, A. Varangrat1, P. Sirivongrangson3, P.A. Mock1, F. van Griensven1
1

HIV Vaccine Trials Network, Hutchinson Cancer Research Center, Seattle, WA, USA; 2San Francisco Department of Public Health, University of California San Francisco, San Francisco, CA, USA; 3Vaccine and Infectious Diseases Division, Hutchinson Research Center, Seattle, WA, USA; 4Ed Wolf Consulting, San Francisco, CA, USA; 5Statistical Center for HIV/AIDS Research and Prevention, FHCRC, Seattle, WA, USA

Thailand MOPH - US CDC Collaboration, Nonthaburi, Thailand; 2Division of HIV/AIDS Prevention, US CDC, Atlanta, GA, USA; 3Bureau of AIDS, TB and STI, Thailand MOPH, Nonthaburi, Thailand

Background: HIV risk reduction counseling (RRC) is an essential component of HIV vaccine trial conduct; strategies to sustain high quality counseling at trial sites are needed. We sought to evaluate the impact of a novel training and support program for site RRC mentors to enhance local RRC quality assurance efforts. Methods: In November 2008, the HVTN implemented an RRC mentor training program for NIAID-sponsored sites and those contracted by Merck, Inc. to conduct the Step trial. Thirty-eight sites designated RRC mentors to participate in a multi-faceted program including an RRC protocol/worksheet, Training of Mentors workshop, monthly webinar/call series, training at HVTN Conferences, personalized and small group consultations with an RRC training expert, an online clearinghouse of RRC resources, and a newsletter. In June 2010, an anonymous internet survey was administered to 203 counseling staff from participating sites as follow-up to a baseline needs assessment survey. Mentor involvement in the HVTN-organized program, types of site-level mentoring activities, and staff perceptions of change in RRC skills were assessed. Results: The overall response rate was 60% (n=121). Approximately half (47%) of respondents were clinicians (physician/nurse). Ongoing RRC mentoring programs were reported by 91% (30/33) of currently active sites; virtually all mentors (97%) participated in the HVTN-organized program. At sites, mentors most frequently facilitated individual or group discussions about challenging counseling issues, provided targeted technical support, and addressed counselor fatigue. Mentors cited time constraints and scheduling problems with staff as the most common barriers to mentoring. A substantial majority (79%) of RRC staff indicated their counseling skills improved over time due to on-site mentoring/training activities with 75% directly attributing an increase in skills to the HVTN program. Conclusion: Survey respondents reinforced that a centralized, multi-faceted training program to support local RRC mentors can facilitate RRC quality assurance efforts at global vaccine trial sites.

Background: To develop clinical research infrastructure and assess hepatitis B (HBV) prevalence, vaccination uptake, and development of vaccine-induced immunity among men who have sex with men (MSM) in Bangkok with high HIV incidence in preparation for HIV prevention trials. Methods: Prospective cohort study, follow-up at 4 monthly intervals for a minimum of 3 years. Study recruitment targeted community based organizations, MSM entertainment venues and the Internet. We asked men to come to the clinic for eligibility evaluation, provide informed consent, complete a behavioral questionnaire using audio-computer-assisted self-interviewing, and undergo medical evaluation and HBV and HIV testing with pre- and post-test counseling. Baseline and 12 month HBV infection status was determined by serology. Those who were susceptible to HBV infection were offered the series of three HBV vaccinations free (ENGERIX-B, 1 mL (20 mcg) given at months 0, 1 and 6). Results: Between April 2006 and January 2008, 1,292 men (mean age 26 years) were enrolled (enrollment rate 98.6%). At baseline, 494 (38.2%) men had immunity from natural infection (anti-HBc +/- anti-HBs), 120 (9.3%) had acute or chronic infection (HBsAg); and 103 (8.0%) had vaccine-induced immunity (isolated antiHBs). Of the remaining 572, 506 (88.5%) consented and received the 1st, 488 (96.4%) received the 2nd, and 453 (89.5%) received the 3rd dose. Of 433 vaccinees that completed 12 months of follow-up, 347 (80.1%) developed vaccine-induced immunity (anti-HBs), 60 (13.9%) developed no antibodies, 17 (3.9%) had evidence of immunity due to natural infection (anti-HBc/antiHBs), 8 (1.8%) showed evidence of acute infection (anti-HBc, no anti-HBs), and 1 (<1%) was indeterminate. Conclusion: High HBV prevalence, high HBV vaccination uptake and high rates of vaccine induced protection were observed in this preparatory cohort of MSM in Bangkok. HBV vaccination may serve as an appropriate model for evaluating and providing access to HIV vaccines among Bangkok MSM.

AIDS Vaccine 2011

127

POSTERS

Posters
P06.13 LB

Topic 06: Clinical Trial Site Challenges P06.14 LB


Title: Screening of Concordant Negative Couples during the Pre-screening Period for a Phase-One HIV Vaccine Trial in Lusaka, Zambia
J.L. Tucker1, W. Kilembe1, M. Goodwin1, E. Hunter1, S. Allen1, F. Priddy2, M. Krebs2, E. Chomba1
1 2

Screening and Enrolment into a Phase I HIV Vaccine Trial In Kigali-Rwanda


R. Bayingana1
1

Project San Francisco/IAVI, Kigali, USA

Background: Pre-screening of potential phase I HIV vaccine volunteers reduces potential screen failures and work load during the screening phase. In a Phase I HIV Vaccine Trial, currently conducted at Projet San Francisco (PSF) in Kigali, Rwanda, we anticipated that by approaching low risk couples for enrollment into HIV vaccine trial and setting pre-screening criteria, we would remove potential barriers to volunteer enrollment and other common causes of enrolment failure. We present the common reasons for volunteer ineligibility despite doing pre-screening. Methods: Between February-April 2011, 206 HIV concordant negative couples were enrolled in the Heterosexual (HT) study and 52 (104) couples were pre-screened out due to not using long term family planning methods (IUD or Implant), or having high HIV risk behaviors, abnormal hematology, chemistry and/or urinalysis, not showing enough willingness to participate, lactation, or had a clinically significant abnormal medical assessment. The remaining 154 couples where at least one partner was potentially eligible for screening underwent; informed consent procedures, medical assessment and screened for hepatitis B and C. Eligible volunteers were invited for enrolment. In an eligiblecouple, only one partner would be enrolled. Results: Out of 154 couples, we screened 137 volunteers. Among these, 61 volunteers were eligible and 76 were ineligible. Of the 76, we had 23 (30%) Hepatitis (16 due to Hepatitis C, 5 due to Hepatitis B and 2 volunteers having co-infection of B and C), 11 (15%) Hepatomegally/splenomegally, 17(22%) other abnormal clinical condition 10 (13%) HIV risky behavior, other 15 reasons were; failure of assessment of understanding of informed consent, lactation, declined enrolment and abnormal urinalysis Conclusion: Our study showed relatively high prevalence of Hepatitis. Among the pre-screening parameters, very few showed up at screening. Therefore if prescreening is well planned and well done, it can reduce the number of screen failures.

Zambia-Emory HIV Research Project, Lusaka, Zambia; International AIDS Vaccine Initiative, New York, NY, USA

Background: In preparation for early phase HIV vaccine trials, ZEHRP maintains a cohort of HIV-concordant negative couples in a prospective observational study (HT). Prior to enrolment in an HIV vaccine study, these individuals are followed up for a period between 6-18 months. Ideal candidates for safety trials are healthy individuals with a low risk for HIV infection or pregnancy. Methods: ZEHRP partners with 15 government clinics where couples are invited to participate in CVCT. Cohabiting concordant negative couples using a long-term contraceptive method are invited to enroll in the HT study. Prior to formal screening for participation in a vaccine study, the researchers performed chart reviews for these individuals to assess study-specific inclusion/ exclusion criteria and a positive attendance record. Results: Of the 206 individuals pre-screened for inclusion in an HIV vaccine trial, 130 were ineligible (64 male; 66 female). The most common reasons for ineligibility were poor attendance record (55) during the HT study, high-risk behavior for HIV infection (36), unwilling/unavailable for duration of study (16), above age limit (16), absence of IUD/implant (9), and syphilis detected (8). Specific reasons for high-risk behavior for HIV infection were multiple sexual partners in the past year (14), partner having multiple sexual partners in the past year (14), frequent, excessive alcohol use (9), trichomonas positive (5), and partner trichomonas positive (5). One participant became infected with HIV during the pre-screening period. Conclusion: Over one quarter (26.7%) of the individuals prescreened demonstrated poor attendance during the prescreening period. Because poor attendance during clinical trials may impair data quality and distort conclusions[1], researchers should employ the pre-screening model whenever possible. Additionally, researchers should prioritize enrolling cohabiting, concordant negative couples. The low rate of seroconversion among our cohort, compared to previous trials that did not account for partner status, might be attributable to this strategy.

POSTERS 128 AIDS Vaccine 2011

Topic 06: Clinical Trial Site Challenges P06.15 LB


High Levels of Willingness to Participate in an HIV Vaccine Trial in Kigali Rwanda
J. Mukamuyango1, E. Karita1, R. Bayingana1, J. Nyombayire1, R. Ingabire1, J. Mukamwezi1, L. Dagna2, P. Fast2, A. Tichacek1, A. Susan1
1

Posters
P06.16 LB
Targeting Concordant HIV Negative Couples as an Effective Recruitment Strategy for Phase 1 HIV Vaccine Trials
J. Nyombayire Mutagisha1, E. Karita1, R. Bayingana1, R. Ingabire1, J. Mukamuyango1, A. Tichacek2, D. Laufer3, H. Thomson3, P. Fast3, S. Allen4
1 Rwanda-Zambia HIV Research Group (RZHRG)-Projet San Francisco,Rwanda, Kigali, Rwanda; 2Rwanda-Zambia HIV Research Group (RZHRG ),Emory University, Atlanta, GA, USA; 3International AIDS Vaccine Initiative, New York, NY, USA; 4Rwanda-Zambia HIV Research Group (RZHRG), Emory University, Atlanta, GA, USA

Rwanda Zambia HIV Research Group/ Projet San Francisco, Kigali, Rwanda; 2International AIDS Vaccine, New York, NY, USA

Background: The concept of participating in clinical trials is still not well understood by African populations with low rates of literacy. In preparation for a phase I HIV vaccine trial, we held information seminars and then conducted an assessment of willingness to participate in the study in a cohort of HIV concordant negative couples in Kigali, Rwanda. Methods: In February-March 2011, HIV negative couples who were enrolled in the Heterosexual Transmission Study were invited for information sessions on HIV vaccine trials. Information sessions included viewing the informed consent video, a group discussion and questions and answers session. After the session, couples were asked verbally if they were willing to participate in the trial, and those interested were given appointment to come back for consenting and screening procedures. Results: A total of 206 HIV concordant negative couples were enrolled. Overall, the literacy level as assessed by the ability to read the local language was 67%. Most prospective volunteers spoke Kinyarwanda and only a very small proportion of study participants could understand French or English (6% of men vs 2% of women). Ninety per cent of the invited couples (186/206) attended an HIV vaccine trial seminar, and the willingness to participate in the study was very high: 179 men (96%) and 174 women (84%) stated that they were interested in participating into the study. At the time of screening, only two couples declared they were no longer interested in this study. Conclusion: This study shows that, despite a low literacy level, when the information about HIV vaccines is provided, willingness to participate in an HIV vaccine trial is very high. This willingness, coupled with the strong reputation of our research center in the community, might contribute to high enrollment rates in HIV vaccine trials.

Background: Recruiting HIV low risk volunteers in phase I HIV vaccine trials can be a challenge in high HIV prevalence settings. Previous studies have shown that HIV incidence among concordant negative couples is very low and in a prior phase I trial with 53 concordant HIV negative couples, no seroconversions were observed in one year of follow-up.Our study aimed at evaluating the effectiveness of couples counseling and testing as an entry point in HIV vaccine trials. Methods: Between August and November 2010, 13 clinics within Kigali were asked to refer concordant negative couples to Projet San Francisco research site in anticipation of a phase I HIV vaccine trial. Couples were either using a long-term family planning method or initiated its use at enrollment. In FebruaryApril 2011, these couples were invited for screening to participate in an HIV vaccine trial. Results: A total of 206 concordant HIV negative couples were recruited. 52 couples (25.2%) were not interested in participating in the trial or were found to be ineligible during the pre-screening phase. No screening procedures were performed in 64 additional couples because the site had reached its enrollment target. In the remaining 90 couples in whom at least one partner was screened, 52 (58%) had at least one partner eligible, whereas in 38 (42%) couples, both partners were ineligible. A total of 45 volunteers (23 men and 22 women) were enrolled in the trial. Conclusion: This study shows that targeting concordant HIV negative couples is an effective strategy to recruit HIV low risk volunteers in phase I vaccine trials. The screening success rate was very high (58%), with nearly a 1:1 ratio of enrolled men and women. Concordant HIV negative couples are at low risk for HIV and should be considered for future safety and immunogenicity trials in high prevalence areas.

AIDS Vaccine 2011

129

POSTERS

Posters
P07.01

Topic 07: Host Genetic Factors P07.02


TRIM5 Does Not Affect SIVmac251 Replication in Vaccinated or Unvaccinated Indian Rhesus Macaques
G. Franchini1, C. Fenizia1, D. Nichols1, S. Cornara1, N. Binello1, M. Vaccari1, P. Pegu1, M. Robert-Guroff2, Z. Ma3, C. Miller3, B. Keele4, D. Venzon5, V. Hirsch6 National Cancer Institute, National Institute of Health, Bethesda, MD, USA; 2Laboratory of Immune Biology of Retroviral Infection Section/NCI/NIH, Bethesda, MD, USA; 3 California National Primate Research Center, USA; 4Cancer Virus Program/ NCI-Frederick, MD, USA; 5Biostatistics and Data Management Section/ NCI, NIH, USA; 6Laboratory of Molecular Biology/NCI/NIH, USA
1

HLA Targeting Efficiency and Its Applications in Predictions of HIV Viral Load and HIV Disease Progression
T. Hertz1, D. Nolan2, I. James2, M. John2, S. Gaudieri2, E. Phillips2, J.C. Huang3, G. Riadi3, S. Mallal2, N. Jojic3 Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Murdoch University, Perth, Australia; 3Microsoft Research, Redmond, WA, USA
1 2

Background: The diversity of HLA binding preferences has been driven by the sequence diversity of short segments of pathogenic proteins, presented by HLA molecules. To identify commonalities in HLA binding preferences, we define a novel measure termed targeting efficiency, which captures the correlation between HLA- binding affinities and the conservation of the targeted proteomic regions. Analysis of targeting efficiencies for 95 HLA Class-I alleles over thousands of human proteins and 52 human viruses indicates that HLA molecules preferentially target conserved regions in these proteomes. We predicted that ranking HLA alleles by their efficiency scores for a given virus would be predictive of the ability to control infection. Methods: We analyzed the effect of efficiency scores for HIV1 proteins on viral load in a study population of 191 HIV-1 clade-B infected, treatment-naive individuals from the Western Australian HIV cohort. We computed HLA targeting efficiency for each individual using their HLA-A and HLA-B alleles, thus approximating the aggregate ability of patient-specific HLA alleles to differentiate between conserved and variable targets. Results: Overall HLA efficiency scores toward HIV-1 proteins were negatively correlated with viral load (ns, p=0.24). However, HLA-B locus efficiency in targeting Gag was more strongly correlated with viral load (r = -0.19, p=0.009), consistent with experimental evidence that HLA-B CTL responses to Gag play a significant role in control of HIV infection. Using multivariate regression to combine efficiency scores of all HIV-1 proteins and proteasomal cleavage, we found that efficiency scores accounted for 7% of variance in viral load (r=0.27, p < 0.0004). We then analyzed the distribution of HLA efficiency scores for HIV-1 Gag and found that protective HLA alleles tend to rank more highly then alleles associated with rapid progression. Conclusion: Ranking HLA alleles or individuals by their efficiency scores is predictive of clinical phenotypes and may assist in HIV vaccine design.

Background: The SIVmac251 macaque model is widely used to evaluate the relative efficacy of HIV vaccine candidates. Thus, the understanding of natural factors that confer resistance to SIVmac251 replication in Rhesus macaques is key to minimize the over estimation of vaccine efficacy. HIV-1 does not infect macaques and the restriction of HIV replication in these old world monkeys is mediated at the post-entry level by the interaction of TRIM5 and the viral capsid (CA) protein. In vitro TRIM5 restriction depends on the dose of SIVmac251 used in the infectivity assay, suggesting the importance of the stoichiometry between the capsid and the TRIM5 proteins. However, TRIM5 in rhesus macaques is highly polymorphic. Recent studies have shown polymorphisms in the B30.2/SPRY domain of rhesus TRIM5 are associated with reduced efficiency of SIVmac251 replication in vivo. Methods: We determined the distribution of polymorphic TRIM5 alleles in a cohort of macaques by DNA sequencing and assessed whether these alleles affected SIVmac251 virus levels and virus induced CD4+T-cell depletion in nave macaques or in macaques immunized with a combination of DNA-SIV/ALVACSIV/gp120 or ALVAC-SIV /gp120 vaccines. The synergistic effect of protective MHClass-I alleles and the role of the dose of virus used in the challenge experiments, was also evaluated. Results: Surprisingly, our results on a cohort of eighty two macaques, forty three vaccinated and thirty nine naive, demonstrated that previously-identified restrictive alleles of TRIM5, did not appear to have a significant effort on SIVmac251 replication in vivo, regardless of prior vaccination for challenge dose. Furthermore, the presence of the protective alleles MamuA01+, B08+, or B017+ did not synergize with vaccination or TRIM5. Conclusion: Polymorphic TRIM5 alleles do not appear to influence the result of vaccination when the SIVmac251 is used in intra-rectal challenge experiments.

POSTERS 130 AIDS Vaccine 2011

Topic 07: Host Genetic Factors P07.03 LB


IRF-1 Polymorphisms and Their Role in Resistance to HIV-1 Infection in Highly Exposed Uninfected Kenyan Female Sex Workers
A. Sivro , R. Su , G. Liu , J. Xie , T.B. Ball , F.A. Plummer
1 1 2 2 3 1 3

Posters
P07.04 LB
Impact of HLA-B*35 Subtype Differences on HIV Outcome in Mexico
C. Juarez-Molina1, S. Avila-Rios2, H. Valenzuela-Ponce2, M. Soto-Nava2, C. Garcia-Morales2, D. Garrido-Rodriguez2, P. Goulder1, G. Reyes-Teran2
1 2

University of Manitoba, Winnipeg, Canada; 2University of Manitoba, Department of Physiology, Winnipeg, Canada; 3 National Microbiology Laboratory, Winnipeg, Canada
Background: Interferon Regulatory factor -1 (IRF-1) is a transcriptional activator of interferon genes and interferon inducible genes and plays a crucial role in host antiviral immunity and HIV replication. DNA sequence variations can cause phenotypic changes by multiple mechanisms, including mRNA splicing and turnover. Previous work has shown association of three polymorphisms in IRF-1 with decreased susceptibility to HIV-1 infection and a reduced likelihood of seroconversion. Peripheral blood mononuclear cells (PBMCs) from patients with protective IRF-1 genotypes exhibited significantly lower basal IRF-1 expression and reduced responsiveness to IFN- stimulation. This study will further characterize the effect of identified polymorphisms on IRF-1 expression and its effect on HIV-1 infection. Methods: Alternative splicing and the functional impact of IRF1 polymorphisms on expression of IRF-1 regulated genes was analysed using the Affymetrix Human Exon 1.0 ST microarray. Quantitative RT-PCR was used to investigate IRF-1 mRNA levels, as well as to confirm the microarray results. IRF-1 protein stability was analysed using Western Blot analysis. Exon splicing and transcript stability assay were performed in order to characterize the link between identified polymorphisms and altered IRF-1 protein levels. Results: Data from this work shows an association of protective IRF-1 polymorphisms with increased expression of exon 7/8 and decreased IRF-1 protein stability. Resulting decrease in IRF-1 protein levels can prevent over-activation of the immune response and hinder HIV-1 replication. Further functional analysis of IRF1 polymorphisms and HIV resistance is ongoing. Conclusion: Individuals with protective IRF-1 genotypes are able to regulate the nature and strength of the immune response to HIV through altered IRF-1 protein stability. It is important to fully characterize the effect of IRF-1 polymorphisms as this will further the understanding of natural resistance to HIV infections and can contribute to the development of novel prophylactic or therapeutic modalities.

Oxford University, Oxford, United Kingdom (Great Britain); CIENI (Center for Research in Infectious Diseases), Mexico City, Mexico

Background: Previous studies have consistently associated HLA-B*35 with rapid disease progression in the context of B clade HIV infection. HLA-B*35 subtypes of the PX group (such as HLAB*3502/03/B*53) have particularly been associated with worse outcome. The mechanisms underlying these observations are not clear. This study focuses on the Mexican HIV epidemic where HLA-B*35 is expressed in approximately one-third of Mexicans. Methods: The study cohort comprised 679 Mexican subjects with chronic HIV B clade infection. All subjects were HLA typed by sequence-based typing (Abbot). Viral load (VL) and CD4+T cell counts were determined by real-time PCR (Abbot) and multiparametric flow cytometry (BD), respectively. Gag and pol genes were amplified from plasma virus as previously described to determine clade of infection. Results: Consistent with previous studies of B clade infected Caucasoid, HLA-B*57 and HLA-B*27 were the most protective alleles. In contrast with earlier work, no significant impact on VL setpoint or absolute CD4+T cell count was observed between the HLA-B*35 PY versus PX groups (p=0.9776 and p=0.2141 respectively). Diverse ranking of HLA-B*35 alleles according to median viral setpoints was observed. HLA-B*3508 was 2nd of 45 HLA-B alleles, while B*3514 was 45th. HLA-B*3508 VL setpoint was significantly lower than HLA-B*3508 negatives (p=0.0211) when the cohort was analyzed excluding previously determined protective alleles B*57 and B*27. HLA-B*3501, a PY allele not considered a risk allele, was significantly associated with higher VL and lower CD4+T cell counts (p=0.0254 and p=0.0037 respectively). Conclusion: These data suggest that certain HLA-B*35 alleles can be protective against HIV disease progression. We observe substantial differences in markers of HIV disease outcome associated with small differences between HLA-B*35 alleles. Defining the mechanisms underlying these differences will facilitate understanding of the mechanisms of immune control or lack of control of HIV, which is of relevance to HIV T-cell vaccine design.

AIDS Vaccine 2011

131

POSTERS

Posters
P07.05 LB

Topic 07: Host Genetic Factors

Specific Structural Interactions Between HLA-B and Epitopic Peptide Differentiate Alleles Associated With HIV Control and Progression
N.G. Holt1, M. Ferber2, S. Wilton1, A. Piechocka-Trocha1, F. Pereyra1, O. Michelin2, B.D. Walker1
1 2

Ragon Institute of MIT, MGH and Harvard, Boston, MA, USA; Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland

Background: HIV controllers maintain low viral loads in the absence of therapy. Recent analysis indicates that specific residues in the peptide binding groove of HLA-B, particularly 67, 70 and 97, predict disease outcome. Methods: Modeller v9.9 software was used to perform homology modeling based on published crystal structures of the HLA-B/peptide complex, focusing initially on HLA B*5701/ B*5801(associated with control) and B*5802 (associated with progression), which differ at positions 94, 95 and 97. Ab initio peptide refinement was used to determine the mode of MHC docking. A mutant peptide library was constructed based on modeling results. We assessed MHC contact residues influencing HIV control and their binding to epitopes. Results: Homology modeling revealed HIV control to be characterized by hydrogen bonds between the epitope and HLA-B*5701 and B*5801 position 97, and by conserved contact residues in the HIV epitope that are more commonly presented in HIV controllers. Furthermore, side chains of the epitope point towards position 97 in controllers, enhancing hydrogen binding. This pattern is markedly different in the context of the B*5802 allele associated with HIV progression, where position 97 is unable to make the strong contacts observed in HIV controllers. Instead, epitope binding is characterized by non-polar van der Waals contacts between HLA-B and hydrophobic side chains. Conclusion: Our data show that strong hydrogen bonding between HLA position 97 and epitopic peptide is characteristic of HLA-B*57 and B*5801 alleles associated with HIV control, whereas weaker, van der Waals forces define B*5802, associated with HIV progression. The presence of significant patterns in MHC binding directly responsible for the strength of the epitope/ HLA-B interaction indicates that a small number of conserved residues are critical in HIV control. Expansion of these studies to other alleles and epitopes is underway to further define HLApeptide parameters associated with effective T cell mediated immune control.

POSTERS 132 AIDS Vaccine 2011

Topic 08: Immune Escape P08.01


Humoral and Cellular Immune Pressures May Converge at the Base of the V3 Loop During Rwandan Subtype A HIV-1 Infection
M.K. Murphy1, L. Yue1, L.J. Hughes1, E. Karita2, S.A. Allen1, E. Cormier3, J. Salazar-Gonzalez4, G.M. Shaw4, J.E. Robinson5, P. Borrow6, P.A. Goepfert4, E. Hunter1, C.A. Derdeyn1 Emory University, Atlanta, GA, USA; 2Projet San Francisco, Kigali, Rwanda; 3International AIDS Vaccine Initiative, London, United Kingdom (Great Britain); 4University of Alabama at Birmingham, Birmingham, AL, USA; 5Tulane University School of Medicine, New Orleans, LA, USA; 6 University of Oxford, Oxford, United Kingdom (Great Britain)
1

Posters
P08.02
The C3V4 Domain Interacts with Structurally Proximal Regions to Mediate Escape from Autologous Neutralizing Antibodies in HIV-1 Subtype C Infection
J.N. Bhiman1, N. Ranchobe1, C.K. Wibmer1, E.S. Gray1, S. Abdool Karim2, C. Williamson3, L. Morris1, P.L. Moore1 National Institute for Communicable Diseases, Johannesburg, South Africa; 2Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa; 3 University of Cape Town, Cape Town, South Africa
1

Background: Despite continual rounds of viral escape, the human immune system deftly targets and eliminates circulating virions during HIV-1 infection. Better understanding how this targeting is accomplished could inform mechanisms to exploit the envelopes vulnerabilities and to design vaccine immunogens that would irreversibly cripple the virus, disallow escape, and force viral clearance. Methods: Along with that of the transmitted founder virus, envelopes from twenty longitudinal neutralizing antibody escape variants were cloned out of plasma from a Rwandan, subtype A-infected HIV1 subject. Envelope sequence analysis revealed three mutational hot spots in gp120: C2, the alpha2 helix, and V5. Single mutations from the escape variants were introduced into the founder virus by site-directed mutagenesis, and resultant envelopes were assayed against autologous plasma and an autologous monoclonal antibody (mAb) to map sites of immune evasion. Peptides representing certain regions of mutation were also fed into the Prediction Algorithm for Proteasomal Cleavages (PAProC). Results: Mutation of either C2 position 293 or alpha2 helix position 337 conferred full escape from autologous plasma at two-months post-infection. Changing alpha2 helix position 340 yielded partial plasma resistance but rendered envelopes sensitive to the autologous mAb. Collectively, these residues responsible for neutralization escape coalesced at a V3-proximal epitope. Of note is the phenomenon that, though the 340 mutation appeared to introduce a humoral vulnerability early during infection, it became fixed in the later viral population. This residue, situated immediately adjacent to the predicted A*0201-restricted TL9 cytotoxic T lymphocyte (CTL) epitope, saw its PAProC score fall from 68.7 to 17.3 when mutated, suggesting that changing this site--though disadvantageous where humoral pressures were concerned--could have mediated CTL escape through a processing mutation. Conclusion: Mechanisms of early subtype A autologous neutralization converge at the base of the V3 loop, and immune escape could be complicated by the viruss requirement to juggle intersecting humoral and cellular pressures.

Background: Most new HIV-1 infections world-wide are caused by subtype C viruses. The C3V4 region of gp120 is a major target for autologous neutralizing antibodies in this subtype. The alpha2-helix, within the C3 region, is more variable, more amphipathic and likely to be more immunogenic in subtype C than in subtype B. This helix contributes to an epitope including the V4 loop and N332 glycan. Methods: Chimeric viruses containing the C3V4 region from a neutralization sensitive virus in a resistant envelope backbone were generated, and tested for neutralization sensitivity using autologous serum. Individuals were classified as C3V4-responders or non-responders depending on whether they developed antibodies to this region. Longitudinal single genome amplicons were obtained from C3V4-responders at 6 and 12 months postinfection. Sequence analysis was used to identify possible escape mutations in the C3V4 and other structurally proximal regions in escaped viruses. The role of these mutations in escape was examined by site-directed mutagenesis. Results: Chimeric viruses were used to identify 13 C3V4responders and 7 non-responders. Escape pathways in five C3V4responders were examined in detail. In CAP244 and CAP136, escape was mediated solely by changes in the C3V4 region. In 3 other individuals, escape from anti-C3V4 responses occurred via interactions between the C3V4 region and structurally proximal regions of the envelope. A glycan shift in V5 and 3 mutations in the alpha2-helix of CAP206 mediated escape. In CAP228 an interaction between residues K290 in the C2 region and Q336 in the alpha2-helix mediated escape, while addition of a glycan in both the C2 and V4 regions of CAP255 resulted in escape. Conclusion: These data confirm that changes solely within the C3V4 region can mediate escape, but in some cases interactions between the C3V4 and other regions in the outer domain are required, enabling multiple routes for escape from anti-C3V4 antibodies.

AIDS Vaccine 2011

133

POSTERS

Posters
P08.03

Topic 08: Immune Escape P08.04


Molecular and Functional Consequences of Immune-Mediated Evolution in HIV-1 Nef During the North American Epidemic
C.J. Brumme1, B. Chan2, T. Markle2, J. Carlson3, E. Martin2, A.Q. Le2, K. Penney2, M. Rahman2, T. Kuang2, P. Nassab2, D.R. Chopera2, M. Markowitz4, M. John5, S. Mallal5, B.D. Walker6, B. Koblin7, K. Mayer8, M.A. Brockman2, A. Poon1, Z.L. Brumme2 BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada; 2Simon Fraser University, Burnaby, Canada; 3 Microsoft Research, Los Angeles, CA, USA; 4Aaron Diamond AIDS Research Centre, New York, NY, USA; 5Murdoch University, Perth, Australia; 6Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 7New York Blood Center, New York, NY, USA; 8Brown University, Providence, RI, USA
1

Control of HIV Viral Replication Mediated by the Synergistic Effects of Unique Combinations of HLA Class I Alleles
Y.E. Wang1, C. Oniangue-Nzda2, A. Schneidewind3, M. Kemper2, E. Mellors2, Y. Qi4, A.D. Gladden2, K.A. Power2, F. Pereyra2, B.D. Walker2, M. Carrington4, T.M. Allen2
1 Dana-Farber Cancer Institute, Boston, MA, USA; 2Ragon Institute of MGH MIT, and Harvard, Boston, MA, USA; 3 Univeresity of Regensburg, Regensburg, Germany; 4SAICFrederick, Inc., NCI-Frederick, Frederick, MD, USA

Background: A major challenge to the development of an effective HIV vaccine is eliciting durable CD8+ T cell responses from which the virus cannot escape. Spontaneous control of HIV replication by particular HLA class I alleles is associated with the targeting of critical regions of the virus which limit effective viral escape due to the impact of mutations on viral fitness. Unique networks of co-evolving or compensatory mutations are known to exist within different HIV proteins, requiring coordinate co-evolution at these sites during viral escape due to structural protein constraints. Because of this requirement of co-evolving sites, we hypothesized that particular pairs of CTL escape mutations may exist that are uniquely deleterious to HIV, revealing novel combination targets for vaccine targeting. Methods: We computationally analyzed mutation patterns of described CTL escape mutations in capsid sequences and crystal structures to identify mutation pairs potentially unable to structurally coexist. We then cross validated identified position pairs with clinical data and performed in vitro viral replication assay on both single and double mutants for four mutation pairs. Results: We identified 20 non-covarying pairs of residues from public datasets (q-value<0.05). Notably, the pairs of HLA alleles selecting for these non-covarying residues were found enriched in individuals controlling HIV. Moreover, while the majority of single mutations tested had minimal effects on viral replication capacity, 3 out of the 4 pairs of mutations significantly impaired viral replication (B52-T280V+B27-SARKLM; B52-T280V+B57A163N; B57-I147L+B14-T303V). Conclusion: These data suggest that the combination of certain HLA class I alleles may enhance the control of HIV replication due to the inability of HIV to effectively escape from coordinated responses against structurally interacting regions of the virus without incurring substantial fitness costs. This study highlights the importance of CTL based vaccines to simultaneously direct immune responses against co-evolving sites that substantially limit the pathways of viral escape.

Background: HLA-restricted CTL responses drive evolution of the highly immunogenic Nef protein, but the extent and functional consequences of population-level adaptation remain unclear. We have used novel historic Nef data to estimate the date and reconstruct the founder virus sequence of the North American epidemic, compare patterns of population-level HLA-associated polymorphisms over time, and assess CD4 downregulation activity of historic and modern Nef sequences. Methods: Plasma HIV-1 RNA Nef sequencing and HLA typing was performed on 241 historic specimens (1979-89). Modern published HLA/HIV datasets served as controls. Timing and sequence reconstruction of the founder Nef was performed using BEAST and HyPhy. HLA-associated polymorphisms were identified using phylogenetically-corrected methods. CD4 downregulation capacity of 52 historic vs. 52 modern Nef sequences was compared using flow cytometric methods. Results: Based on Nef sequences, the most recent common ancestor of the North American epidemic was dated to 1965. The consensus of the reconstructed founder Nef sequence differed from 2004 subtype B consensus at codons 15, 22, 51 and 178, while additional sites remained ambiguous in the reconstruction. Patterns and statistical strengths of HLAassociated polymorphisms remained generally consistent over time (e.g. A*24-associated Y135F, B*07-R71K, B*08-K94Q and B*57-H116N ranked among the strongest in historic and modern cohorts); however, a small number of polymorphisms were identified as candidates for population-level accumulation. Functional assessment of Nef revealed a modest yet statistically significant increase in CD4 downregulation capacity over time (median 0.93 vs. 1.00 in historic vs. modern sequences; p=0.005). Conclusion: Modest population-level immune adaptation in Nef, potentially leading to modest increases in CD4 downregulation capacity, may have occurred in North America since 1979. However, the relatively high similarity between the estimated founder and modern consensus B, and the observation that CTL escape patterns have remained largely consistent over time, support Nef as a suitable target for vaccine consideration.

POSTERS 134 AIDS Vaccine 2011

Topic 09: Innate Immunity P09.01


Sex Differences in the TLR-Mediated IFN-Alpha Responses Are Associated with Higher Expression of Interferon-Stimulated Genes in HIV-1-Infected Women
J. Chang1, M. Woods1, R.J. Lindsay1, E.S. Chan2, E. Doyle1, M. Griesbeck1, R.J. Bosch2, M. Altfeld1
1

Posters
P09.02
Differential Regulation of Various TLR Pathways in Acute and Chronic HIV-1 Infection
J. Chang1, A. Lacas1, R.J. Lindsay1, E. Doyle1, K. Axten1, B.D. Walker1, F. Pereyra1, T. Allen1, M. Altfeld1
1

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2Harvard School of Public Health, Boston, MA, USA

Background: Previously we have shown that stimulation of plasmacytoid dendritic cells (pDCs) using HIV-1-derived TLR7 ligands induces a significantly stronger IFN-alpha response in females than in males, and higher level of CD8+ T-cell activation (Meier/Chang Nat Med 2009), which has been shown consistently to correlate with HIV-1 disease progression to AIDS. Direct consequences of sex differences in IFN-alpha secretion in response to HIV-1 on T-cell activation still needs to be assessed. Methods: Samples from 38 treatment naive HIV-1 infected subjects (19 females and 19 males) from ACTG 384 were sorted using flow cytometry into dendritic cell and CD4+ and CD8+ T-cell subsets. Cells were lysed and mRNA expression of TLR pathway, IFN-alpha pathway and interferon stimulated genes (ISGs) were analyzed by utilizing the new Nanostring technology. This method allows for amplification-free examination of gene expression using molecular barcodes and can be used on much smaller cell numbers than microarray analysis. Results: We observed no significant difference in the expression of ISGs in CD8+ T-cells between females and males. However, after adjusting for HIV-1 viral load we observed significantly higher levels of ISG expression in CD8+ T-cells isolated from females as compared to males (ISG15 p<0.05 and MX1 p<0.05 using ANCOVA). This is consistent with our previously published finding showing that females had significantly higher percentage of activated (CD38+HLA-DR+) CD8+ T-cells than males after adjusting for viral load. Conclusion: These data show that sex differences in TLR-mediated activation of pDCs is echoed in CD8+ T-cells with higher ISG expression in HIV-1-infected women compared to men at a given viral load. This provides in vivo evidence that sex differences in IFN-alpha responses directly affect CD8+ T-cell immune activation in HIV-1 chronically infected individuals, potentially resulting in the described faster HIV-1 disease progression in females compared to males for the same viral load.

Background: Innate immune cells can sense pathogens through Toll-like receptors. Innate immune responses to pathogenderived TLR ligands in HIV-1 infection have been suggested to be universally dysregulated, with reduced responsiveness to some TLR ligands due to chronic activation-induced anergy and elevated responses to other TLR ligands associated with enhanced general immune activation. Here we systematically compared responses to TLR ligands in individuals with acute HIV infection, chronic untreated viremic, chronic untreated controlled (elite controllers), and chronic treated HIV-1 infection. Methods: Cryopreserved PBMCs from 39 HIV-1 infected subjects and 11 HIV-1 negative individuals were stimulated with CL097 (TLR7/8 ligand), ODN2216 (TLR9 ligand), and heat killed Listeria monocytogenes (TLR2 ligand). TLR-specific responses by monocytes, mDCs and pDCs (APCs) were quantified by intracellular cytokine staining using flow cytometry. Results: Different stages of HIV-1 infection were associated with either reduced or enhanced responsiveness of APCs to TLR ligands. Responses to TLR9 ligands were reduced in all HIV-1infected individuals. In contrast we did not observe an HIV-1associated reduction in responses to TLR7 or TLR8 ligands, but significantly enhanced levels of responses to TLR8 ligands by monocytes and mDCs that were positively associated with HIV-1 viral load (p<0.01 for both). Conclusion: While initial studies had suggested that HIV-1 infection induces a general impairment in the APC responses to TLR ligands, more recent studies have suggested that the responsiveness of APCs to TLR7/8 ligands is preserved in infected individuals (OBrien JCI 2011). Our data are in line with these recent findings, and furthermore demonstrate that responses by monocytes and DCs to pathogen-derived TLRs are differentially regulated in different stages of infection.

AIDS Vaccine 2011

135

POSTERS

Posters
P09.03

Topic 09: Innate Immunity P09.04


Human and Rhesus Macaque TRIM5 Can Bind HIV-1 Cores and Stabilize CA Disassembly
M. Briones1, S. Tekeste2, S. Chow2
1

CCR5 Expression on Peripheral Blood Cells Differs Between South African Caucasian and African Populations
A.C. Picton , S. Shalekoff , M. Paximadis , C.T. Tiemessen
1 2 2 1 1

National Institute for Communicable Diseases and WITS University, Johannesburg, South Africa; 2National Institute for Communicable Diseases, Johannesburg, South Africa

AIDS Research Alliance, Los Angeles, CA, USA; 2University of California, Los Angeles, Los Angeles, CA, USA

Background: CCR5 is pivotal in determining an individuals susceptibility to HIV-1 infection and rate of disease progression. To establish if population-based differences exist in cell surface expression of CCR5 we evaluated the extent of CCR5 expression across all peripheral blood cell types in individuals from two populations, South African Africans (SAA) and South African Caucasians (SAC). Methods: CCR5 expression on peripheral cell populations was determined in EDTA-anticoagulated whole blood of 22 SAA and 32 SAC HIV uninfected individuals (age and gender matched), using a CCR5 antibody conjugated to PE at a ratio of 1:1, thereby allowing for CCR5 quantification as the mean number of CCR5 molecules per cell in addition to the percentage of CCR5-expressing cells within a cell subset. CCR5 expression was determined for B-cells, monocytes, and T-cell and natural killer (NK) cell subsets. Results: Differences in CCR5 expression, both in number of CCR5 molecules per cell and percentage of CCR5-expressing cells, were observed between the two study groups, within all cell subsets. Most notably, the percentage of CCR5+ cells were significantly lower in SAC compared to SAA individuals (P<0.001) among NKcell subsets (CD16+; CD56+ and CD56dim) whereas CCR5 intensity of expression was significantly higher in SAC compared to SAA individuals in CCR5+CD8+ T-cell and CCR5+ NK-cell subsets (CD16+; CD56+ and CD56dim) (all P<0.05). These relationships were maintained post-exclusion of CCR5delta32 heterozygous individuals (n=7) from the SAC dataset. Conclusion: CCR5 expression differs significantly between SAA and SAC individuals. These differences can be attributed to factors other than CCR5delta32. It remains to be determined which genetic factors are contributing to these expression differences. Observed differences in CCR5 expression levels have implications not only for HIV-1 infection, but also for T-cell immunity where CCR5 has also been shown to play a key role.

Background: After membrane fusion with a target cell, the human immunodeficiency virus type 1 (HIV-1) core enters the cytoplasm where uncoating occurs, an important, yet poorly understood event. The cone-shaped core is composed of a hexameric lattice of the viral capsid protein (CA), which disassembles during uncoating. The host restriction factor TRIM5 targets the incoming viral core, resulting in a block to infection, possibly by accelerating or disrupting uncoating. Restriction occurs in a species-specific manner, whereby HIV-1 is weakly restricted by human TRIM5 (TRIM5-Hu), but potently restricted by rhesus macaque TRIM5 (TRIM5-Rh). TRIM5 restriction is presumed to be dependent on the ability to recognize the viral core via CA affinity, but the overall mechanism is not completely understood. Methods: Through the use of isolated cores, endogenous sources of TRIM5, over-expressed TRIM5 in 293T cells and recombinant proteins, the recognition requirements were investigated. In addition, the effects of TRIM5 on uncoating were examined using an in vitro core disassembly assay. Results: Both over-expressed TRIM5-Hu and TRIM5-Rh were able to recognize and co-sediment with isolated cores through a sucrose cushion. Furthermore, endogenous sources of TRIM5 and recombinant proteins were examined and confirmed that both have affinity for the viral core. Using an in vitro core disassembly assay, uncoating was not accelerated; rather core disassembly was stabilized in the presence of endogenous TRIM5 and cell lysates over-expressing either TRIM5. However; the presence of recombinant TRIM5-Rh alone was able to stabilize core disassembly, while recombinant TRIM5-Hu was not. Conclusion: The results suggest that CA-binding is required, but not sufficient for TRIM5-mediated restriction of viral infection. The data supports a model in which the restriction occurs in a step-wise manner and is dependent on additional and yet unidentified cellular factors, with the generation of a stable viral core-TRIM5 complex representing an intermediate in the TRIM5 restriction process.

POSTERS 136 AIDS Vaccine 2011

Topic 09: Innate Immunity P09.05


High Frequencies of Polyfunctional CD8+ Natural Killer Cells in Chronic HIV-1 Infection Are Associated with Slower Progression to AIDS
H. Hong1, M. Jkel2, F. Ahmad2, J.M. Eberhard2, N. Bhatnagar2, M. Ballmaier2, R.E. Schmidt2, D. Meyer-Olson2
1 2

Posters
P09.06
Reduced Expression of LEDGF/p75, an Essential Host Factor for HIV Integration, in HIV Exposed Seronegative Individuals
K. Mous1, W. Jennes2, M. Camara3, M. Seydi3, G. Daneau2, S. Mboup3, L. Kestens2, X. Van Ostade1 University of Antwerp, Wilrijk, Belgium; 2Institute of Tropical Medicine, Antwerp, Belgium; 3Cheikh Anta Diop University, Dakar, Senegal
1

Harvard Medical School, Southborough, MA, USA; Medizinische Hochschule Hannover, Hannover, Germany

Background: Large genotypic studies suggested a protective role of natural killer (NK) cells in HIV infection. The aim of our study was to define correlates of NK cell subpopulations and their functional profiles with slower HIV disease progression. Methods: Associations of lymphocyte subpopulations with HIV disease progression was assessed in an observational study in 116 untreated HIV-seropositive donors with a CD4+ T helper cell count > 500 cells/l who were followed for 4 years. Phenotypic and functional analyses of NK cells from 63 individuals, including 37 untreated and 11 treated HIV-seropositive and 15 uninfected controls subjects, were performed by polychromatic flow cytometry. Statistical analysis included unpaired t test, One-way ANOVA followed by Tukey test, Log-rank test and Pearson analysis. Results: High absolute numbers and percentages of CD8+CD3cells were associated with significantly slower HIV-disease progression (P = 0.048 and P = 0.0074, respectively). The majority of CD8+CD3- cells were CD56-expressing NK cells. Frequencies of CD8+ NK cells inversely correlated with HIV viral loads (r = -0.5, P = 0.0012) and loss of CD8+ NK cells could be restored in HIV-patients after antiretroviral treatment. Analysis of IFN--, TNF--, MIP-1- and CD107a-responses in NK cells after IL-12, IL-15- and K562-stimulation revealed a more polyfunctional profile amongst CD8-expressing NK cells in comparison with their CD8-negative counterpart. Higher frequencies of granzyme B- and perforin-expressing cells were detected amongst CD8+ NK cells as compared to NK cells devoid of CD8 but no significant differences were observed in surface expression of NKG2A, NKG2D, CD62L, CD69, CD2 and CD94. Conclusion: These data provide evidence for a strong inverse relationship between numbers of highly functional CD8+ NK cells and HIV-disease progression. It is tempting to speculate that innate immune responses can contribute to viral control and disease outcome, which might have implications for future vaccine design strategies.

Background: HIV-1 replication in host cells depends on a delicate balance between intrinsic immune factors limiting viral dissemination like apolipoprotein B mRNA-editing catalytic polypeptide-like 3G (APOBEC3G), tripartite motif 5alpha (TRIM5alpha) and tetherin (BST-2), and cellular co-factors required for virus propagation like lens epithelium-derived growth factor (LEDGF/p75). We analyzed whether the expression of these host proteins contributes to the phenomenon of HIV-1 resistance. Methods: Twenty-three HIV-1 exposed seronegative subjects (HESN), 23 healthy controls (HC), and 45 HIV-1 infected subjects of which 9 were therapy nave (HIV-nave) and 36 were treated with antiretroviral therapy (HIV-ARV) were enrolled from a cohort of HIV-negative, HIV-discordant and HIV-concordant Senegalese couples. We used real-time quantitative polymerase chain reaction (qPCR) to measure mRNA expression in PBMC. Protein expression levels were investigated in CD4+ lymphocytes and CD14+ monocytes by indirect fluorescent antibody staining and flow cytometry. Results: LEDGF/p75 protein levels were significantly lower in CD4+ lymphocytes of HESN than in those of HC. This observation was especially true for HESN subjects with higher frequencies of unprotected sexual exposure. For APOBEC3G, TRIM5alpha, and tetherin, no differences were found between HESN and HC. HIV-nave patients generally expressed higher mRNA and protein levels for the four HIV-1 related host factors than HC. In particular for tetherin, increased protein expression levels correlated positively with the enhanced levels of immune activation in these HIV-nave patients. Conclusion: Our observations suggest that reduced LEDGF/ p75 protein expression may contribute to the phenomenon of HIV-1 resistance. Increased expression levels of APOBEC3G, TRIM5alpha, tetherin, and LEDGF/p75 in HIV patients are a likely consequence of the increased levels of immune activation. Understanding the role of HIV-1 related host cell proteins in HIV1 susceptibility may be of great interest for the development of future therapies for the prevention of HIV infection.

AIDS Vaccine 2011

137

POSTERS

Posters
P09.07

Topic 09: Innate Immunity P09.08


ABSTRACT WITHDRAWN

Different Cytokine Profiles Induced by HIV-1 and HIV-1-Derived TLR Ligands in Monocytes and mDCs
R.P. Simmons1, E. Groden1, J.J. Chang1, R. Lindsay1, L. Fadda1, J.D. Lifson2, K. Lane1, K. Axten1, E. Rosenberg3, T. Allen1, M. Altfeld1
1

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2AIDS and Cancer Virus Program, National Cancer Institute, Frederick, MD, USA; 3Massachusetts General Hospital, Boston, MA, USA

Background: Shortly following HIV-1 acquisition, a host of plasma cytokines and chemokines rise sharply. The goal of this study was to characterize the cytokine profile in response to HIV1 viremia and to identify the different pathways leading to HIV1-induced cytokine production. Methods: We evaluated plasma levels of 19 cytokines from a single-arm trial evaluating combination antiretroviral therapy started during acute HIV-1 infection followed by treatment interruption in 14 participants, using a multiplex Luminex bead assay. Sources of cytokine production were identified in vitro following stimulation of PBMCs from healthy individuals with HIV-1 and HIV-1-derived TLR ligands by intracellular cytokine staining (ICS). Results: The plasma cytokine most significantly associated with HIV-1 viral load among the 19 analytes was the chemokine IP10 (CXCL10) (R2 = 0.4, p=< 0.0001). Incubation of PBMCs with TLR8 ligands (CL097 and gag1166 ssRNA), AT-2 inactivated HIV1, and NL43 virus in vitro resulted in a significant production of IP-10 (p = 0.001). IP-10 was produced mainly by mDCs and monocytes in response to AT-2 HIV-1 (average of 59% and 76%, respectively) and NL43 virus (average of 74% and 91%, respectively). In contrast, incubation with TLR8 ligands was associated with increased production of TNF by monocytes and mDCs and only slight increase in IP-10 production. Thus, the cytokine profile of monocytes and mDCs stimulated with whole viruses (AT-2 HIV-1 and NL43) differed from that observed in response to TLR8 ligands alone, suggesting the involvement of additional receptors than TLR8 in the recognition of HIV-1 by these cells. Conclusion: Monocytes and mDCs produce significant amounts of IP-10 in response to stimulation with HIV-1, while stimulation with HIV-1-derived TLR8-ligands results in the preferential production of other pro-inflammatory cytokines, such as TNF. Further studies are needed to gain a better understanding of the pathways that lead to the activation of innate immune cells by HIV-1.

POSTERS 138 AIDS Vaccine 2011

Topic 09: Innate Immunity P09.09


Innate Immune Responses Among Highly Exposed Seronegative Individuals to a Live Attenuated Influenza Vaccine as a Model for Mucosal Viral Infection
I.A. Onyango1, A. Meyers2, Y. Keynan2, J. Oyugi3, J. Kimani1, F. Plummer2, J. Ochanda 1, T.B. Blake2 University of Nairobi, Nairobi, Kenya; 2University of Manitoba, Winnipeg, Canada; 3Univesity of Nairobi, Nairobi, Kenya
1

Posters
P09.10
Effects of a Therapeutic Dendritic Cell-Based Vaccine for HIV-1 Infection on Innate Immunity
J. Pea1, M. Fras2, L. Castro-Orgaz1, H. Arberas3, R. Gonzlez1, F. Garca3, T. Gallart3, A. C. Guardo3, J. Gatell3, M. Plana3
1

Immunology Service, Maimonides Institute for Biomedical Research, UC, Crdoba, Spain; 2Immunology Service, Maimonides Institute for Biomedical Research, UniC, Crdoba, Spain; 3IDIBAPS-FCRB, Hospital Clnic-UB, Barcelona, Spain

Background: Increasing evidence suggests that innate responses are key determinants of the outcome of infection, particularly the role of Toll-like receptors (TLRs) which have recently been shown to be important in early host defense and instruction of adaptive response. In this study, we aimed to understand the earliest event in mucosally transmitted HIV-1 infection, since exposure to HIV in these women is exclusively through the genital mucosa. To better understand the correlates of mucosal protection against HIV infection, a viral challenge was used. The model consists of a live attenuated influenza vaccine administered intranasally to stimulate the mucosal system. Blood was collected prior to (day 0) and following vaccination day 7 and 30. We hypothesize that the HIV resistant individuals would have heightened innate immune responses compared with HIV susceptible individuals. Methods: The study population was drawn from the Majengo sex worker cohort, Nairobi, Kenya comprising 2 groups: Long term HIV exposed but uninfected (HESN/ HIV resistant); short term HIV exposed but uninfected (New Negatives/ susceptible) commercial sex workers. Peripheral blood mononuclear cells (PBMCs) were isolated from the study participants. After incubation with agonists specific for TLR9 (CpG ODN) and TLR7/8 (ssRNA40), in vitro production of cytokine and chemokine were quantified using multiplex assay on Luminex. Results: Stimulation of PBMCs with TLR 7, 8, 9 ligands led to a significant elevated levels in IL-2 at baseline (p=0.0159) and following vaccination day 30 (p=0.0079) in HESN compared to HIV susceptible individuals. Elevated chemokine IFN- -induced Protein-10 (IP-10) were observed HESN compared to HIV susceptible individuals 30 days post vaccination (p=0.0317). Conclusion: HIV-resistant women had stronger IL-2, IFN- and IP-10 responses to TLR ligands compared to HIV-susceptible. This may point to an important protective role of innate mechanisms in influencing susceptibility to HIV-1 infection, and has a potential for augmenting prevention or therapeutic interventions.

Background: NK cells dysfunction contributes to the progression of HIV-1 infection, but no data exist concerning their role in response to HIV-1 therapeutic vaccines. Here we present the results obtained analysing NK cells subpopulations and their regulatory receptors in HIV-1 chronic infected patients enrolled in a MD-DC based therapeutic HIV vaccine trial. Methods: 23 chronic HIV-1 infected patients with baseline CD4+ T cells above 450 cells/mm3, BVL above 10,000 copies/mL and off HAART for at least the last 2 years were randomized (1:1) to be immunized every 2 weeks (w0, w2 and w4) with 3 doses of autologous MD-DCs either pulsed ex vivo with whole autologous heat-inactivated HIV-1or non-pulsed. Immunological parameters were followed-up until w48 The NK cell subpopulations CD56bright, CD56dim and CD56neg and their regulatory receptors CD85j/ILT-2, CD94, NKG2A and NKG2C were measured by flow cytometry. Results: The levels of the different NK subpopulations analysed in HIV-1-immunized patients and placebo indicate that levels of the CD56neg NK cell subpopulation rose in immunized patients from week 24 until the end of the time period studied but no significant differences were found for CD56bright and CD56dim NK cell subpopulations. When the expression of NK regulatory receptors was analysed, no significant differences were observed for NK subsets for CD94-NKG2A and CD94- NKG2C receptors. However, the percentage of NK subpopulations expressing CD85j/ ILT-2 was significantly lower in CD56dim and CD56bright NK cell subpopulations in immunized patients than in placebo individuals. Conclusion: The increase in the levels CD56neg NK subpopulation with high capacity to produce chemokines involved in HIV1 suppression as well as the decrease in the expression of the inhibitory receptor CD85j/ILT-2 in both NK CD56bright and NK CD56dim subpopulation, fact that could facilitate cytolytic and secretory functions, in immunized patients should be of clinical relevance after a MC-DC based therapeutic vaccine.

AIDS Vaccine 2011

139

POSTERS

Posters
P09.11

Topic 09: Innate Immunity P09.12


Correlation Between AIDS Pathology and the Biased Nucleotide Composition of Lentiviruses
N. Vabret1, M. Bailly-Bechet2, A. Lepelley1, V. Najburg1, M. Mller-Trutwin1, O. Schwartz1, B. Verrier3, F. Tangy1 Institut Pasteur, Paris, France; 2Universit Lyon I, Lyon, France; 3IBCP, Lyon, France
1

A Role for the KIR2DL3 Receptor in Mother-toInfant HIV-1 Transmission


M. Paximadis1, G. Minevich2, R. Winchester2, D.B. Schramm1, G.E. Gray3, G.G. Sherman4, A.H. Coovadia5, L. Kuhn6, C.T. Tiemessen1 National Institute for Communicable Diseases, Johannesburg, South Africa; 2College of Physicians and Surgeons, Columbia University, USA; 3Chris Hani Baragwanath Hospital, Soweto, Johannesburg, South Africa; 4University of the Witwatersrand Medical School, Johannesburg, South Africa; 5University of the Witwatersrand, Empilweni Clinic, Johannesburg, South Africa; 6Mailman School of Public Health, Columbia University,, USA
1

Background: Natural killer cells are thought to be involved in attenuation of HIV-1 disease, primarily through recognition by killer-cell immunoglobulin-like receptors (KIRs) of specific HLA Class I molecules. The influence of KIR and HLA on maternal-toinfant transmission however has not been studied. We investigated the role of the weakly inhibitory KIR receptor, KIR2DL3 and its HLA-C ligand (C1) on perinatal HIV-1 transmission. Methods: KIR genes and HLA Class I C* alleles were determined in Black South African HIV-1 infected mothers (224) and their infants (72 infected and 150 exposed-uninfected). Analyses involved comparisons of total groups, stratification on the basis of maternal nevirapine (NVP)-exposure, and adjustment for maternal viral load (MVL). Results: KIR2DL3 homozygosity alone and in combination with HLA-C allotype heterozygosity (C1C2) was elevated in intrapartum (IP)-transmitting mothers compared to non-transmitting mothers (P=0.034 and P=0.01 respectively), and post-MVL correction (P=0.033 and P=0.027, respectively). In infants, KIR2DL3+C1 as well as homozygosity for KIR2DL3 with C1C2, were both found to be underrepresented in infected infants compared to exposed-uninfected infants in the total group (P=0.06 and P=0.038, respectively) and in the sub-group of infants whose mothers received NVP (P=0.007 and P=0.03, respectively). These associations were stronger post-MVL adjustment (total group: P=0.02 and P=0.009, respectively; NVP group: P=0.004 and P=0.02, respectively). Because of the apparent conflicting associations of KIR2DL3/KIR2DL3+C1C2 with regard to maternal IP-transmission and infant susceptibility, the level of concordance between mothers and infants harbouring this genotype was investigated. Results point to a protective effect for this genotype in the infant only when there is discordance (i.e. mother lacking genotype) and that this protection is likely to be in the context of IP transmission. Conclusion: These results indicate a role for KIR2DL3 in perinatal HIV-1 transmission and highlight the complexities in studying the influence of host genes in both maternal transmission and infant susceptibility.

Background: AIDS results from generalized immune activation after years of chronic HIV replication. However, most African primate species naturally infected with lentiviruses remain healthy. Addressing this issue is crucial for understanding AIDS pathogenesis. Because the genomes of lentiviruses present a particularly biased nucleotide composition compared to that of their primate hosts, we wondered whether host recognition of biased viral nucleic acids could induce hyper-responsiveness to HIV and AIDS pathogenesis. Methods: We compared the nucleotide composition (A/C/ G/T frequencies) of pathogenic and non-pathogenic primate lentiviruses with that of their hosts for every infection described in the literature. We measured the nucleotide divergence by computing the Chi-2 distance between the A/C/G/T frequencies in the complete viral sequence and the coding sequences of each host organism. Results: We found that primate lentiviruses having the most divergent nucleotide composition compared to their hosts induce AIDS, whereas less divergent lentiviruses cause non-pathogenic infections. Similarly, the relative pathogenicity of HIV-1 subtypes correlates with their nucleotide divergence to the human genome. To understand this observation at a molecular level, we investigated the ability of HIV-1 RNA fragments to stimulate in vitro the synthesis of type I interferon (IFN-I). We found that the nucleotide divergence of RNA fragments strongly correlates with type-I IFN activity. Based on these observations, we designed a large-scale, nucleotide-optimized SIV sequence derived from the pathogenic clone SIVmac239. We produced the corresponding artificial virus in lymphocyte cell line and analysed its capacity to induce type-I IFN in different cellular models. This synthetic virus presented the same replicative capacity than WT virus and a reduced ability to induce type-I IFN. Conclusion: Overall, these data suggest for the first time a direct link between the nucleotide composition of lentiviruses and their pathogenicity. They also describe the synthesis a novel artificial SIV harbouring an attenuated pathogenic potential.

POSTERS 140 AIDS Vaccine 2011

Topic 09: Innate Immunity P09.13


Dysregulation of Tim-3 Expression on NK Cells in Chronic HIV-1 Infection
S. Jost1, U.Y. Moreno-Nieves1, J. Reardon1, I. Toth1, A. Piechocka-Trocha1, G. Alter1, M. Altfeld1, M.M. Addo1
1

Posters
P09.14
Early Phosphorylation Events Induced in Dendritic Cells by the HIV Derived TLR8 Ligand ssRNA40
A.J. Leslie1, T. Pichulik1, E. Khatamzas1, A. Mayer1, A. McMichael1, A. Simmons1
1

Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA

University of Oxford, Oxford, United Kingdom (Great Britain)

Background: Chronic exposure to HIV-1 antigens leads to T-cell exhaustion and an impaired virus-specific T-cell response, which correlates with increased expression of several immunoregulatory molecules including T-cell immunoglobulin domain and mucin domain 3 (Tim-3). Interestingly, NK cells constitutively express high levels of this receptor and Tim-3 has been identified as a gene that is significantly up-regulated following NK cell activation (Hanna et al., 2004). Moreover, up-regulation of Tim3 on NK cells has recently been associated with reduced antiviral properties (Ju et al., 2010). However, the effect of chronic HIV-1 infection on Tim-3 expression on NK cells and on Tim-3mediated NK cell function has not been studied yet. Methods: We examined Tim-3-expression on NK cells by flow cytometry in HIV-1 elite controllers (HIV-VL <50 copies/ml), viremic controllers (HIV-VL <2000 copies/ml), individuals with treated and untreated chronic progressive HIV-1 infection and HIV negative control subjects. Results: Tim-3 expression was high on NK cells from HIV negative individuals (median percentage 75.8, range 60.7-83.9). However, proportions of Tim-3+ NK cells were significantly decreased in all HIV-infected individuals compared to healthy subjects (p=0.0001), with a marked down-regulation of this receptor at the surface of NK cells in elite (p=0.002) and viremic controllers (p=0.04). Conclusion: Chronic exposure to HIV-1 leads to significantly decreased levels of Tim-3 expression on NK cells, in striking contrast to its upregulation on exhausted T-cells in untreated HIV-1 infection. This may suggest distinct Tim-3 signaling pathways in NK and T cells. Further investigation is warranted to evaluate the impact of Tim-3 and its ligand galectin-9 on the regulation of NK cell function in the context of HIV-1 infection. Defining the role of NK cells receptors in the control of HIV-1 will offer novel therapeutic targets to manipulate to improve future HIV-1 vaccine strategies.

Background: Dendritic cells (DC) are among the first cell-type encountered by HIV, and their interaction plays a crucial role in subsequent viral spread. Studies show that HIV infection leads to abnormal DC maturation and an impaired interferon response. However, HIV contains at least one TLR7/8 ligand, which can induce maturation and cytokine production, and stimulation of these receptors has recently been found to be crucial for viral replication in DCs. This work aims to understand how HIV is able to subvert TLR signalling in DCs, allowing for replication but mitigating the anti-viral interferon response. Methods: Monocyte-derived dendritic cells were stimulated with HIV-TLR8 ligand ssRNA40, synthetic TLR8 ligand R848 or a control for 10-minutes and assayed for protein phosphorylation changes using a novel method. Briefly, this involved enrichment of phosphorylated proteins, fractionation by isoelectricfocusing and tagged mass-spectrometry to identify differentially phosphorylated proteins. Results were confirmed by Western blot. Results: Over 100 proteins were found to be differentially phosphorylated after 10minutes stimulation with ssRNA40 and R848. Pathway analysis shows these to group into known immune signalling networks. Interestingly there are striking differences between ssRNA40 and R848; with ssRNA40 biased towards a more regulatory/inhibitory outcome. Amongst the currently confirmed targets, this is characterized by profound dephosphorylation of ERK1/2, compared to the expected phosphorylation observed with R848; and phosphorylation of the inhibitory molecule IRAK-M. QPCR analysis of cytokine profiles and TLR expression also suggests a difference in the signalling induced by ssRNA40 and Conclusion: This methodology provides a powerful and unbiased method of looking at early signalling events induced by TLRstimulation. Target confirmation is on-going, but initial data indicate that the HIV-1 TLR8-ligand ssRNA40 may induce a distinct inhibitory signal compared to R848. This raises interesting questions as to how HIV modulates TLR8 signalling in DCs to allow for replication but diminish the anti-viral immune response

AIDS Vaccine 2011

141

POSTERS

Posters
P09.15

Topic 09: Innate Immunity P09.16


Ontogeny, Breadth, and Specificity of Circulating ADCC-Mediating Antibodies in HIV-1 Infected Individuals
J. Pollara1, L. Morris2, N. Meguid1, J. Pickeral1, J. Hoxie3, J. Kappes4, C. Ochsenbauer4, K. Soderberg1, G. Tomaras1, B. Haynes1, D. Montefiori1, G. Ferrari for the CHAVI and CAVD-CAVIMC Teams1
1

Mapping of ADCC Activity: The A32 Conformational C1 Epitope is a Dominant Target for ADCC Antibodies in Chronically HIV-1 Infected Subjects
G. Ferrari , J. Pollara , D. Kozink , T. Harms , M. Drinker , S. Freel1, M.A. Moody1, G.D. Tomaras1, C. Ochsenbauer2, J.C. Kappes2, G.M. Shaw2, J.A. Hoxie3, J.E. Robinson4, D.C. Montefiori1, B.F. Haynes for the CHAVI and CAVD-VIMC Teams1
1 1 1 1 1

Duke University Medical Center, Durham, NC, USA; University of Alabama at Birmingham, Birmingham, AL, USA; 3 University of Pennsylvania, Philadelphia, PA, USA; 4Tulane University, New Orleans, LA, USA
1 2

Duke University, Durham, NC, USA; 2National Institute for Communicable Diseases, Johannesburg, South Africa; 3 University of Pennsylvania, Philadelphia, PA, USA; 4 University of Alabama at Birmingham, Birmingham, AL, USA
Background: ADCC is a contributing determinant of the immune responses controlling HIV-1 replication and may constitute a relevant function of vaccine-induced antibodies. We sought to determine the ontogeny, breadth, and specificity of ADCCmediating antibodies in plasmas collected from HIV-1 acutely and chronically infected individuals. Methods: Longitudinal plasma samples were collected from 3 acutely infected individuals from 1 month to over 1 year post transmission. Cross-sectional plasma samples were collected from 9 chronically infected individuals who represented the top 3% of broadly neutralizing antibody responses (Elite Neutralizers = EN), 14 individuals lacking broad neutralizing capabilities (Tier 1 neutralizers =T1N), and 13 individuals maintaining virus load below 2,000 RNA copies/mL (Controllers). We measured the ability of these plasmas to mediate ADCC responses directed against target cells that were either infected with HIV-1, or coated with recombinant gp120s representing subtypes B, C, and E.

Background: Among non-neutralizing HIV-1 envelope antibodies, those capable of mediating antibody dependent cellular cytotoxicity (ADCC) activity have been postulated to be important for control of HIV-1 infection. However the precise targets of serum ADCC antibody are poorly characterized. We utilized the human nonneutralizing CD4 induced (CD4i) mAb A32 and monomeric A32Fab, that recognize a conformational C1-C4 region to map ADCC Ab responses. Methods: We utilized flow cytometry to evaluate the expression of the conformational epitope recognized by the A32 mAb on cells infected with transmitted/founder and laboratory adapted HIV-1 isolates. The ADCC activity of the A32, 17b, 12b, PG9, and 2G12 mAb were evaluated using HIV-1-infected cells. In order to map the ADCC specificity of the Ab responses elicited by HIV-1 infection, we used the A32-Fab to pretreat our target cells and prevent binding of A32-like Ab present in the samples collected from 40 chronically infected donors of whom 13 were Controllers. Results: We observed that mAb A32 epitope was expressed on the surface of HIV-1 infected CD4+ T cells earlier than the CD4inducible (CD4i) epitope bound by mAb 17b and the gp120 carbohydrate epitope bound by mAb 2G12. The mAb A32 was able to mediate ADCC activity 2-4 fold higher than any other mAb at peak of activity. After pre-incubating the target cells with the monomeric A32-Fab, we observed a range of inhibitory activity for the peak of the response from 19% to 90% in samples collected from all subjects (averagestandard deviation = 5624). Moreover, A32-like ADCC responses represented the majority (>50 inhibition) of the peak response in 69% Controllers (n=13), 57% Tier 1 neutralizers (n=14); 22% Elite neutralizers (n=9). Conclusion: Our data suggest that CD4i Ab, and in particular those recognizing the A32 conformational epitope, play a major role in mediating ADCC responses in the course of HIV-1 infection.

Results: Cross-clade ADCC-mediating antibody responses were detectable within 4 months of transmission. In chronic infection, Area Under the Curve (AUC) analysis indicated that ADCC activity directed against HIV-infected cells was higher in the Controllers (AUC=1349) and EN (AUC=6915) compared to T1N (AUC=48). We observed a positive correlation between ADCC activities directed against gp120-coated and HIV-1 infected targets in EN and Controllers, but not in the T1N. Among the ENs, there was no association between titers of gp120 binding antibodies and ADCC activity (p=0.38, R2=0.11) and a negative correlation between ADCC activity and gp41-biding antibody titers (p=0.05, R2=0.49). Conclusion: Our data suggest that cross-clade ADCC activity directed against HIV-infected cells arises early after infection, but is underrepresented in the majority of chronically infected individuals compared to Controllers. Broadly reactive antibodies specific for conserved regions of gp120 are likely responsible for ADCC responses directed against infected cells, and may represent an important target for efficacious HIV vaccines.

POSTERS 142 AIDS Vaccine 2011

Topic 10: Mucosal Immunity P10.01


Intranasal Co-Administration of IL-12 plus CTB in DNA-MVA Mucosal Schemes Enhanced Systemic and Mucosal Cellular Immune Responses Against HIV-1 Env
C.A. Maeto1, A.M. Rodrguez1, J. Falivene1, M.F. Pascutti1, M.M. Gherardi1
1

Posters
P10.02
CD107a Expression Rather than Polyfunction by HIV-Specific T Cells in the Female Genital Tract Was Associated with Protection from HIV Shedding
A. Bere1, W.A. Burgers1, L. Denny1, J. Pasmore1
1

Centro Nacional de Referencia para el SIDA, Buenos Aires, Argentina

University of Cape Town, Cape Town, South Africa

Background: The major route of HIV transmission is through mucosal tissues. Therefore, designing immunization regimes aimed to induce mucosal immune responses is needed. The aim of this study was to analyze the activity of IL-12 alone or in combination with the cholera toxin B subunit (CTB), applied in DNA-prime/MVA-boost intranasal immunizations. Methods: Balb/c mice were intranasally immunized with both vectors expressing HIV-1 EnvB. Two doses of DNA-IL-12 were applied (50g or 100g) in the presence or not of 10g of CTB applied at prime and booster doses. Groups receiving CTB, complete cholera toxin (CT) or non-adjuvants (control) were included. Immune responses were evaluated 14 or 30 days after immunization. Results: Both DNA-IL-12 doses generated similar responses, even more the minor one plus CTB generated the highest response, showing a synergistic effect for both adjuvants. Co-inoculated IL-12+CTB (G4) produced the highest specific TCD8+ immune response (by IFN-gamma ELISPOT), detected in the spleen (up to a seven-fold increment), as well as in regional (cervical) and genital (iliac) lymph nodes (GLNs), and more importantly in genital tract mucosa (GT). IL-2 secreting cells were two-fold superior in G4 compared to control group in spleen (p=0.0321), GLNs (p=0.005) and GT. A higher proportion of cytotoxic and polyfunctionality cells were detected in spleen and GT. At memory phase, we found that in the G4 IFN-gamma and IL-2 secreting cells were two to three-fold superior in both systemic (spleen, p=0.001) as well as in mucosal compartments (GLNs). Finally, IL-12 and CTB also enhanced gp120 Abs levels (serum IgGs and IgA in vaginal washings). Conclusion: Here we demonstrated that IL-12 cytokine in combination with CTB generated a cooperative adjuvant effect on the cellular and humoral IR against Env antigen applied in DNA-MVA intranasal immunizations. These results are important due to the need to improve mucosal vaccine strategies against HIV.

Background: HIV-specific T cells are present in the female genital tract of chronically HIV-infected individuals. The aim of this study was to compare the polyfunctional ability of HIV-specific mucosal T cell responses in the female genital tract with those in blood and their relationship with HIV clinical status and genital HIV shedding. Methods: Cervical cytobrush-derived and blood-derived T cells were obtained from 16 chronically HIV-infected women. Cervical CD3+ T cells were expanded with Dynal anti-CD3/CD28 expander beads for 14 days to increase T cell numbers for further analysis. Polychromatic flow cytometry was used to simultaneously evaluate four T cell functions (CD107a, IFN-, TNF-, and MIP1) and compare them to HIV clinical status. Results: TIn response to HIV, CD107a was found to be the dominant response of CD8 T cells in both the cervix and in blood. Expression of CD107a in both compartments was negatively associated with plasma viral load. In the genital tract, CD107a expression by genital tract CD8 T cells was associated with protection from HIV shedding. While the majority of Gag-specific T cell responses in the female genital tract and blood were monofunctional, low frequencies of HIV Gag-specific polyfunctional CD8 T cells were detected at the cervix and was found to correlate significantly with polyfunctional frequencies in blood. The polyfunctionality of blood Gag-specific CD8 T cells was associated with clinical status (blood CD4 count and plasma viral load). No association was found between polyfunctional responses at the cervix and clinical status or genital HIV shedding. Conclusion: HIV Gag-specific cervical T cells were largely monofunctional and CD107a appears to be associated with better control of HIV shedding in the genital tract. Polyfunctional T cells were detected in women with high blood CD4 count and low plasma viral load and these did not protect from HIV genital shedding.

AIDS Vaccine 2011

143

POSTERS

Posters
P10.03

Topic 10: Mucosal Immunity P10.04


TSLP: A Potential New Mucosal Adjuvant for Intranasal Immunisation with gp140
G.A. Van Roey1, M. Arias2, J.S. Tregoning2, R.J. Shattock2
1 St Georges University of London, London, United Kingdom (Great Britain); 2Imperial College, London, United Kingdom (Great Britain)

Evaluation of Mucosal Immunization Routes for Induction of Vaginal Humoral Immunity in Macaques
R. Veazey , A.A. Siddiqui , V. Buffa , L. Fischetti , D. King , R.J. Shattock2
1 2 2 2 2

Tulane National Primate Research Center, Covington, LA, USA; 2Imperial College London, London, United Kingdom (Great Britain)
1

Background: The predominant route for sexual HIV transmission is via genital and rectal mucosal surfaces. The development of a successful vaccine against HIV is likely to require induction of strong and long-lasting humoral immune responses at the mucosal portals of virus entry. Hence, the design of a vaccine strategy able to induce mucosal antibodies may be crucial to providing immune protection. This study has assessed how the route of vaccination and adjuvant used modifies mucosal responses to model antigens in macaques. Methods: Female Rhesus macaques (n=4 per group) were immunized three times, either intranasally, sublingually or intrarectally with model antigens (KLH, OVA or B-gal). Immunizations were performed every four weeks and animals were treated with medroxyprogesterone 4 weeks prior to first administration. Four different adjuvants were assessed per route of administration (including R848 (resiquimod), chitosan, TSLP, Poly-IC, and Pam3CSK4). Results: Intrarectal and sublingual immunization failed to induce humoral responses in macaques. Intranasal immunization induced potent responses when R848 was used as an adjuvant, but not with the other adjuvants tested. Intranasal immunization with R848 induced potent and systemic immune responses including localized induction of IgA (nasal and vaginal). In depth safety studies to assess local and systemic cytokine and inflammatory responses to nasal application of R848 and characterization of nasal cellular influx indicate that this approach was well tolerated. No adverse events to nasal application of R848 were detected. Conclusion: These data demonstrate that intranasal immunization with R848 as an adjuvant provided potent systemic and localized specific immune responses, both IgA and IgG. This route was much more effective in induction of specific vaginal humoral responses than sublingual or intrarectal routes of immunization. Ongoing studies will assess whether similar results can be obtained for gp140.

Background: The induction of strong and long-lasting humoral immune responses at the mucosal portal of virus entry is most likely required for a successful HIV vaccine. There is an urgent need of a safe and potent mucosal adjuvant for human use that could induce these immune responses. Our previous findings showed that intranasal (i.n.) immunisation with HIV-1-gp140 and TSLP as adjuvant induces antibody responses in serum and vagina. To further explore the adjuvant effect of TSLP, we assessed cellular immune responses to HIV-1 gp140 after i.n. immunisation and their impact on the longevity of systemic and mucosal humoral responses. Responses were compared to those induced using the potent mucosal adjuvant cholera toxin (CT). Methods: Mice were immunized three times i.n. in the course of 9 weeks. gp140-specific IgA and IgG responses in serum and vagina were assessed. Splenocytes and cells from the female genital tract were assessed for the presence of antibody secreting cells. Spleens were assessed for T-cell proliferative responses and T-cell-specific cytokine production. Results: TSLP induced strong humoral responses, comparable to those seen with CT. Interestingly, vaginal gp140-specific IgA and IgG were still high 6 months after the last boost. In addition, TSLP enhanced T-cell proliferative responses to gp140 i.n, but the percentage of CD4+ and CD8+ T-cells producing cytokines induced by TSLP after i.n. immunisation was not different from that induced by gp140 alone suggesting the adjuvant effects are predominantly mediated through enhancement of B cell responses in keeping with its known effect on B cell proliferation and differentiation. Conclusion: Our results show that TSLP induces high and longlasting humoral immune responses both systemically and at the vaginal mucosa. The ability of TSLP to induce high and long lasting gp140-specific vagina IgA following nasal immunisation suggests a novel adjuvant strategy for preventative HIV vaccines.

POSTERS 144 AIDS Vaccine 2011

Topic 10: Mucosal Immunity P10.05


DNA Prime-rAd5 Boost Vaccine Applied to the Tonsils Does Not Protect Against Intrarectal Challenge Despite Strong Immune Responses in Rhesus Macaques
A. Duerr , H. Park , A. Sylwester , P. Kozlowski , N. Coombes , A. Nogueron2, J. Vogt1, G. Spies1, L. Picker2, M. Axthelm4
1 2 2 3 2 1

Posters
P10.06
Microneedle Mediated Intradermal Delivery of Adjuvanted Recombinant HIV-1 CN54gp140 Effectively Primes Mucosal Boost Inoculations
A. Pattani1, P.F. McKay2, R.F. Donnelly1, M.J. Garland1, K. Migalska1, C.M. Cassidy1, R. Malcolm1, R.J. Shattock3, R.M. Curran1
1

HIV Vaccine Trials Network, Seattle, WA, USA; 2Vaccine and Gene Therapy Institute/ OHSU, Beaverton, OR, USA; 3 Louisiana State University, New Orleans, LA, USA; 4Oregon National Primate Research Center, Beaverton, OR, USA
Background: Induction of potent mucosal immunity would be desirable for HIV vaccines. However, the effect of mucosal versus parenteral vaccination routes on immunogenicity and protective efficacy remains unclear. Tonsil immunization, which may elicit genital and GI tract immunity, is particularly interesting for HIV vaccines. Methods: Mamu-A*01(+)/B*17(-)/B*08(-) rhesus macaques were immunized intramuscularly (IM) or submucosally over the tonsils (IT). 8 animals per group were primed with plasmid DNA encoding SIVmac239 Gag, Pol, Env, Vif at months 0, 1, 2 and boosted at 6 months with rAd5 vectors expressing Gag, Pol, Env, Vif-Vpr-Vpx. Cellular responses in blood and bronchoalveolar lavage (BAL) were evaluated by ICS and tetramer binding assays. Antibodies in plasma and secretions were quantitated by ELISA. 6 months after the last vaccination, animals were rectally challenged with escalating low doses of SIVmac239. Results: IT and IM immunization induced similar levels of plasma SIV-specific antibodies. IT vaccination induced greater mucosal IgA responses, primarily in the respiratory tract with lower magnitude rectal responses. Peak CD8 T cell responses in PBMC, BAL and, to a lesser extent, colon were higher in the IT vs. IM group. All unvaccinated control animals were infected after 7 challenges. All IM and IT animals were infected after 11 and 12 challenges, respectively. Peak viral load was significantly lower in vaccinated animals than controls but did not differ by vaccination route. Anamnestic humoral and cellular responses also did not differ by vaccination route, with rapid development in blood and respiratory tract but delayed and low magnitude responses in the rectum. Conclusion: Tonsillar immunization with DNA/rAd5 appeared not to confer benefit over IM immunization with respect to rectal SIV acquisition or viremia, despite superior mucosal responses. Possible explanations include use of a neutralization-resistant challenge virus, poor vaccine-induced responses at the site of challenge or use of a vaccine that induces primarily CD8 responses.

Queens University of Belfast, Belfast, United Kingdom (Great Britain); 2St.Georges University of London/Imperial College London, United Kingdom (Great Britain); 3Imperial College London, United Kingdom (Great Britain)

Background: Microneedles have previously been shown to enhance transdermal delivery of drugs and macromolecules. Their ability to effectively penetrate the dermis over a relatively large surface area offers potential use as minimally-invasive and painless replacements for more conventional parenteral vaccine regimens. In this study we compared the immune responses to HIV CN54gp140 using microneedles alone or a microneedle prime followed by mucosal boosting with monophosphoryl lipid A adjuvanted protein. Methods: Polymeric microneedle arrays that dissolve after piercing the skin were fabricated from poly(methylvinylether/ maleic acid) (Gantrez AN139) using laser-engineered silicone micromould templates. Three groups of 8 BALB/c mice were primed using microneedles containing CN54gp140 and monophosphoryl lipid A. Three boosts were performed at two week intervals, using either the same microneedle formulation or vaginal or nasal administration of an antigen/adjuvant solution. Another group received four subcutaneous immunisations. Mice were sampled (serum and vaginal wash) prior to each vaccination and two weeks post final immunization. Antigen-specific IgG and IgA production was assessed in the sera and mucosal lavage samples by quantitative ELISA. Splenocytes were harvested at necropsy and analysed for lymphocyte proliferation. Results: CN54gp140-specific serum IgG and lymphocyte proliferation responses were elicited in the subcutaneous immunization group and in those animals that were primed using microneedle delivery and boosted mucosally via the nasal route. Only the microneedle prime / intranasal boost group mounted a robust IgA response. In this group, IgG1 and IgG2a subtype quantification revealed a bias toward IgG1. Conclusion: We have demonstrated that using a novel microneedle device to prime immunity followed by an intranasal boost elicits significant antigen-specific immune responses. This regimen generated similar serum IgG levels to the subcutaneous inoculations, but in contrast IgA was significantly elevated. This vaccination modality also induced a bias toward the IgG1 a subtype suggesting a Th2 skewing of the immune response phenotype.

AIDS Vaccine 2011

145

POSTERS

Posters
P10.07 LB

Topic 10: Mucosal Immunity P10.08 LB


A Phase1 Clinical Trial to Evaluate the Safety, Mucosal and Innate Immunity of Adenovirus Type 26 HIV-1 Vaccine in Healthy, HIV-1 Uninfected Adults
L.R. Baden1, J. Liu2, H. Li2, S. Walsh1, J. Johnson1, D. Milner1, M. Seaman2, K. Krause1, E. Swan3, R. Tucker1, M. Weijtens4, M. Pau4, R. Dolin2, D.H. Barouch2
1

Characterization of the Immune Response in the Gut Mucosa and Peripheral Blood in Acute HIVInfected Thai Individuals
A. Schuetz1, Y. Phuang-Ngren1, V. Assawadarachai1, R. Trichavaroj1, S. Jongrakthaitae1, N. Phanuphak2, S. Pinyakorn2, R. Rerknimitr3, W. Ridtitid3, D. Suttichom2, N. Chomchey2, N. Michael4, P. Phanuphak2, M. de Souza1, J. Kim4, J. Ananworanich2
1

USAMC-AFRIMS, Bangkok, Thailand; 2SEARCH/The Thai Red Cross AIDS Research Centre, Bangkok, Thailand; 3Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; 4US Military HIV Research Program, Rockville, MD, USA
Background: Mucosal surfaces are the major entry portal for HIV. The majority of CD4+ T cells are lost during acute HIV-infection, with mucosal compartments being most severely affected. Understanding the mucosal immune response in early HIVinfection is crucial for the development of an effective vaccine or treatment. Methods: Thirty Thais with acute HIV-infection were enrolled and subsequently received antiretroviral therapy (ART). Prior to (baseline) and 6 months following ART initiation, mucosal mononuclear cells (MMC) isolated from sigmoid biopsies and peripheral blood mononuclear cells (PBMC) were analyzed for their immunological phenotype and HIV-specific immune response using multi-parameter flow cytometry. Results: At baseline, the frequency of CD4+ T cells in MMC and PBMC was comparable to healthy individuals with a median of 48% and 54% respectively, with CD4+ central memory T cells in MMC (56%) and nave CD4+ T cells in PBMC (53%) being the dominant population. There were no changes observed under ART. In contrast, the frequency of CD4+CCR5+ T cells in the MMC at baseline was lower (median 38%) compared to published data for healthy individuals (60-80%) with a trend to increase under ART. At baseline, the frequency of activated CD8+ T cells (HLA-DR+/CD38+) in MMC and PBMC was significantly increased at Fiebig III compared to Fiebig I (p=0.01 and p=0.03, respectively) while a significant reduction in the activation status was observed following 6 months of ART (MMC: p=0.01; PBMC: p=0.003). The HIV-specific immune response in PBMC was mainly CD8-mediated with 5/11 (45%) individuals responding at baseline against Gag, Env and/or Pol compared to 1/11 (10%) responding at baseline in the CD4 compartment. Conclusion: In acute HIV-infection immunological changes in the mucosa include depletion of CD4+CCR5+ T cells and activation of CD8 T cells occurring as early as Fiebig I to III with evidence for trend reversal with ART.

Brigham and Womens Hospital, Boston, MA, USA; 2Beth Israel Deaconess Medical Center, Boston, MA, USA; 3DAIDS, NIAID, NIH, Bethesda, MD, USA; 4Crucell, Netherlands

Background: HIV is largely acquired via mucosal contact. Characterizing mucosal immune responses is a priority. In addition, the STEP study has raised important questions regarding potential interactions of preexisting immunity to a recombinant Ad5 vectored HIV vaccine on risk of acquisition to HIV infection. Methods: This is a placebo-controlled RCT to evaluate the safety and innate and mucosal immune responses of a single intramuscular dose of an HIV-1 recombinant Clade A env (Ad26. ENVA.01) vaccine at a dose of 5x 1010 VP in Ad26 seronegative (n=16) and seropositive (n=8) subjects. Blood samples for standard and innate immune assessments were collected on days 1, 3, 7, 14 and 28. Mucosal samples including rectal wicks and biopsies were collected at baseline and days 14 and 168. Blood and colorectal biopsy specimens were processed fresh within 2 hours of collection for multiparameter flow cytometry. Results: 21/24 (88%) subjects have been enrolled (14/16 in Ad26 seronegative and 7/8 in Ad26 seropositive groups). 52% are < 30 years old, 52% female, and 39% non-Caucasian and 12% Hispanic/Latino. No vaccine related SAEs have occurred, and no significant reactogenicity has been observed. Preliminary data from the first 12 Ad26 seronegative subjects showed EnvAspecific CD8+ T lymphocyte responses in PBMC and colorectal mucosa following vaccination. No increase in Ki67+ cellular activation, or in CCR5+ expression in total or vector specific CD4+ T lymphocytes were seen in PBMC and colorectal mucosa samples obtained before and after vaccination. Conclusion: Ad26-EnvA vaccination is generally well tolerated and elicits both peripheral and mucosal EnvA-specific cellular immune responses. In Ad26 seronegative subjects, there is no evidence to date of increased total or vector-specific Ki67+ or CCR5+ CD4+ T lymphocytes in colorectal mucosa following Ad26-EnvA vaccination. Detailed characterization of mucosal antigen- and vector-specific immune responses in both Ad26 seronegative and seropositive subjects is ongoing.

POSTERS 146 AIDS Vaccine 2011

Topic 10: Mucosal Immunity P10.09 LB


Intramuscular Priming with Replicating Vaccinia Vector and Intranasal Boost With HIV gp140 Trimer Elicit Strong Mucosal and Systematic Immune Response
S. Wang , F. Liu , Y.Y. Liu , R. Wang , Z. Qi , Y. Liu , Y. Shao
1 1 1 1 1 1 1

Posters
P10.10 LB
Targeting HIV Gag p24 to Non-human Primate Dendritic Cells via DCIR and Langerin Evoke Potent and Long-lasting Antibody Responses
R. Le Grand1, S. Zurawski2, N. Dereuddre-Bosquet1, V. Contreras1, I. Mangeot1, F. Martinon1, S. Oh2, J. Banchereau2, G. Zurawski2, Y. Levy3 CEA, Fontenay-aux-Roses, France; 2Baylor Institute for Immunology Research, Dallas, TX, USA; 3INSERM U955, Crtil, France
1

Chinese Center for Disease Control and Prevention, Beijing, China


1

Background: Mucosal immunity at the portal of entry plays a crucial role in protection against HIV infection. However, there is no ideal approach to induce robust mucosal immune responses against HIV-1. Here we described a prime /boost immunization strategy which can induce effective mucosal and systemic immune response. Methods: HIV-1 CN54 structural genes carrying replicating Tiantan vaccinia (rTV) and Listeria (Lm) and mammalian cell expressed gp140 trimer were used to immunize BALB/c mice systematically (im, sc and ip) or mucosally(in, ig). Three main prime boost strategies were investigated: rTV + gp140 trimer; rTV + Lm + gp140 trimer and rTV + Lm/ gp140 trimer. Oil in water, Cholera toxin B (CTB) and Flagellin were used as test adjuvant. Blood, spleen, saliva and vaginal lavage were collected and envspecific IgG and IgA antibody (ELISA) as well as T cell response (Elispot) were determined. Results: Oil in water adjuvant is shown to induce better both serum and mucosal IgA than CTB or Flagellin. rTV is a better priming and gp140 or Lm/gp140 is a better boosting agents. rTV systematic, not mucosal, prime and gp140 or Lm/gp140 mucosal, not systematic, boost, induce the highest serum and mucosal IgA and systematic responses than other immunization strategies. The mice immunized with rTV (im)+ gp140 (in) induced very high and balanced HIV-1 env specific antibody titer both in serum (IgG >150,000, IgA >15,000) and at mucosal level (saliva IgA >600, vaginal IgA >500). rTV (im)+ gp140 /Lm (in) immunization induced a slightly higher IgG in serum and detectable T cellular response but lower IgA antibodies in serum and at mucosal level. Conclusion: Replicating viral vector systematic priming and mucosal protein boosts can induce strong and balanced mucosal and systematic antibody response to HIV-1. This immunization strategy is valuable in AIDS vaccine development.

Background: Targeting antigens to dendritic cells (DC) via antiDC receptor antibody-antigen fusion proteins offers a novel paradigm for evoking potent humoral and cellular responses. Methods: DCIR is a receptor broadly expressed on antigenpresenting cells and Langerin is expressed on Langerhans cells. Chimeric hIgG4 antibodies cross-reacting to Cynomolgus macaque receptors were engineered with C-terminal H-chain fusions to HIV Gag p24 or H1N1 HA. A total of 52 macaques were vaccinated i.d. with 250 g of anti-DCIR-Gag p24, antiLangerin-Gag p24, control human IgG4-Gag p24, Gag p24 adsorbed onto nanoparticles, or Gag p24 protein (at a molar equivalent dose), with and without 250 g poly I:C. Vaccinations were repeated at weeks 6 and 15. Serum and BAL samples were analyzed for antibodies against Gag p24 and HA. T cell responses were measured by IFN/IL-2ELISpot and cytokine production by antigen-specific proliferating cells. Results: In non-adjuvanted nave animals serum anti-Gag p24 titers were detectable 2 weeks after priming, but only in the antiDCIR-Gag p24 and anti-Langerin-Gag p24 groups. Antibody titers increased substantially after both the 1st (3.60.6 and 3.80.5 log titers for anti-DCIR-Gag and anti-Langerin-Gag groups, respectively) and 2nd boosts in the DC-targeting groups and were long-lasting, while low-minimal responses were detected in the control groups. Poly I:C increased the kinetics and magnitude of the responses in the anti-DCIR-Gag p24 (4.70.1) and human IgG4-Gag p24 groups (4.60.1) evoked low responses in the Gag p24 group (3.20.2), and had minimal impact on responses in anti-Langerin-Gag p24 group (4.00.5). HIV Gag and H1N1 HA specific T cell responses from anti-DC receptor targeting were significantly improved in animals previously primed with Gag nanoparticles and PR8 H1N1 infection. High recall HI serum responses were oberved with anti-DC-HA Conclusion: This study provides the context for development of vaccines based on HIV Env fused to DC-targeting antibodies.

AIDS Vaccine 2011

147

POSTERS

Posters
P11.01

Topic 11: Novel Immunogens/Inserts P11.02


A Brain-Derived HIV-1 Envelope Protein Clone with Utility as an Immunogen
R.J. Center1, J. Tan1, J. Sterjovski2, L.R. Gray2, C. Siebentritt1, M.J. Churchill2, S.M. Reddy1, P.A. Ramsland2, P.R. Gorry2, D.F. Purcell1
1

Systemic and Mucosal Vaccination of Rabbits Using Envelope Glycoproteins Derived from HIV-1 Pre-Seroconversion Strains as Novel Immunogens
S. Reddy , R. Center , K. Suzuki , B. Chua , D. Jackson , P. Gorry3, A. Kelleher4, D. Purcell1
1 1 2 1 1

The University of Melbourne, Melbourne, Australia; 2Centre for Immunology, St. Vincents Hospital,, Sydney, Australia; 3 Center for Virology, Burnet Institute,, Melbourne, Australia; 4 National Centre in HIV Epidemiology and Clinical Research, Sydney, Australia
1

University of Melbourne, Parkville, Australia; 2Burnet Institute, Prahran, Australia

Background: Novel immunogens capable of eliciting quality humoral responses could serve as potential HIV-1 vaccine candidates. Envelope glycoproteins (Env) from HIV-1 preseroconversion (PSC) strains are antigenically distinct from late stage Env due to absence of immune selection pressure early during infection and may function as novel immunogens by eliciting effective antibody responses. Methods: Functional PSC env gp160 clones derived from patient samples were used to generate gp140 clones which were engineered into cell lines to produce soluble gp140. Rabbits were vaccinated with gp140 DNA plasmids and gp140 protein trimers in a double DNA prime triple protein boost regime testing two different PSC Env antigens. Both mucosal and systemic vaccinations were performed - intranasal and intramuscular routes for DNA arm followed by intranasal and subcutaneous routes for protein arm. Novel lipopeptide R4Pam2Cys adjuvant was used and additionally Montanide was tested for use with proteins in systemic route. Samples collected include blood, vaginal washes, fecal pellets and nasal washes. Quantitative antibody responses were measured as IgG, IgA and IgM antibody titres and qualitative antibody responses were measured as neutralization against HIV-1 pseudoviruses in TZM-bl cells. Results: IgG, IgA and IgM antibodies were detected in sera and mucosal samples. Sera demonstrated up to log106.5 IgG titres with high avidity and up to 99% neutralization against MN pseudoviruses. The group receiving Montanide systemically had enhanced humoral response as evidenced by antibody titres and neutralization in comparison to group receiving Pam2Cys and sustained it possibly due to depot effect. All samples are further being tested for neutralization against a panel of HIV-1 Conclusion: Thus PSC Env antigens are capable of eliciting quality humoral responses both systemically and mucosally in rabbits especially neutralizing antibodies and can serve as novel vaccine immunogens. Challenge studies in non-human primates will further help evaluate the usefulness of these novel immunogens as vaccine candidates.

Background: HIV Env properties including high sensitivity to neutralization by the CD4 binding site (CD4bs)-targeting neutralizing antibody (NAb) b12 and low CD4 dependency have been associated with brain tropism. These observations suggest that brain-derived Env may be in a relatively open conformation, with increased exposure of functionally conserved domains. We hypothesized that when used as an immunogen this may translate to the generation of broader and more potent NAb responses targeting these conserved domains. Methods: Brain-derived Env clones were sequenced by standard methods. Mice were immunized with an Env DNA prime/trimeric protein boost regimen. Sera were assessed for Env-binding antibodies by ELISA and virus neutralization was measured using a pseudotyped reporter assay. Modeling was performed using the structure of the b12-bound gp120 core. Glycans were manipulated by site directed mutagenesis and b12 binding was assessed by ELISA. Results: Initial screening of brain-derived Env clones indicated that they had a stronger affinity for b12 and sCD4 than spleenderived clones from the same cohort. Immunization with Env of one brain-derived clone elicited better NAb responses than other clones tested. Sequencing of this clone revealed that conserved glycans at residues 197 and 386, which have been shown in previous studies to protect the CD4bs, were absent. Predictably, restoration of the glycan at residue 386 reduced b12 binding. Unexpectedly, restoration of the glycan at residue 197 enhanced binding, as did removal of the glycan at residue 362. Modeling identified other glycans that may affect exposure of the CD4bs due to their proximity to this domain. Conclusion: We have identified a brain-derived Env clone that appears to have utility as an immunogen. The glycans of this clone have a significant but not entirely predictable role in CD4bs exposure. Artificial manipulation of the glycans may allow further enhancement of immunogenicity by increasing CD4bs exposure while still maintaining structural stability.

POSTERS 148 AIDS Vaccine 2011

Topic 11: Novel Immunogens/Inserts P11.03


Generation of HIV-1 Virus-like Particles Expressing Different HIV-1 Glycoproteins
M. Visciano3, L. Diomede1, M. Tagliamonte3, M. Tornesello3, V. Asti1, M. Bomsel2, F.M. Buonaguro3, L. Lopalco1, L. Buonaguro3
1 San Raffaele Scientific Institute, Milan, Italy; 2Institut Cochin, Universit Paris Descartes, Paris, France; 3National Cancer Institute, Naples, Italy

Posters
P11.04
Evaluation of the Immune Response Elicited by Retroviral Vectors Based on HIV-1 Genome in Asymptomatic HIV+ Chronic Individuals
A.C. Guardo1, C. lvarez-Fernndez1, J. Garca-Prez2, F. Garca3, J. Gatell3, J. Alcam2, H. Arberas1, S. Snchez-Palomino1, M. Plana1
1

Background: Elicitation of a potent and broadly neutralizing antibody response is the main goal of an effective preventive HIV1 vaccine. It has been shown by us and others that the expression of env glycoproteins on the surface of particulate structures, such as Virus-Like Particles (VLPs), could be a more efficient strategy to deliver conformational epitopes to the immune system. Methods: To this aim, VLPs expressing native HIV env gp140 or gp41 glycoproteins have been produced in insect cells using a baculovirus expression system and characterized for appropriate protein expression. VLP-bound HIV gp140 glycoprotein showed the appropriate expression and trimeric conformation. Immunogenicity studies have been performed in BALB/C mice by intra-peritoneal administration and sera from immunized mice have been tested in ELISA assays, for their reactivity with HIV specific antigens, as well as in ex vivo neutralization assay. Results: Sera from immunized animals showed a high reactivity with individual HIV proteins expressed in VLPs. Results of TZMbl based neutralization assay show that combined sera from animals independently immunized with gp140- or full-lengthgp41-expressing VLPs have an additive/synergistic effect in the neutralization activity of HIV pseudoviruses. Conclusion: In conclusion, novel VLPs expressing different HIV env glycoproteins with native trimeric conformation have been generated, showing the induction of effective antibody response with neutralization activity in TZM-bl neutralization assay. These results confirm the effectiveness of VLPs as presentation and delivery system for conformational proteins and show the improved neutralization activity upon the combination of antisera elicited by different HIV envelope antigens, suggesting the possibility of broadening the spectrum of viral epitopes targeted by immune response.

IDIBAPS-FCRB-Hospital Clnic, Barcelona, Spain; 2AIDS Immunopathology Unit, National Center of Microbiology, ISCIII, Madrid, Spain; 3Infectious Diseases Unit, Hospital Clnic, IDIBAPS, Barcelona, Spain
Background: The development of HIV vaccines is an urgent priority and there is need to generate reagents that can be used to screen HIV-1-specific responses. Whole inactivated viral particles have been successfully used as vaccines for some viruses, but procedures historically used for inactivation can denature viral proteins reducing their immunogenicity and could be potentially dangerous for the patients. We have evaluated the utility of retrotranscriptase defective virions (NL4-3/DRT) as a potential immunogen. Methods: IFN- ELISpot assay has been developed to evaluate specific T cells response against different types of NL4-3/DRT virions (including aldrithiol-2 treated, X4 and R5 tropic particles) using cryopreserved PBMC from up to 68 asymptomatic HIV+ individuals. Alternatively NL4-3 wild type inactivated virions (aldrithiol-2 treated; WT+AT-2) and pools of overlapping peptides (15 mer) encompassing the HIV-1 Gag and Nef regions have been used as a control. Extracellular stainings have been also carried out to measure immunological features. Results: Preliminary immunogenicity studies showed that positive ELISpot responses were detected in 38/68 (56%) and 16/68 (24%) of individuals tested with RT or WT+AT-2 virions respectively. The magnitude of the total responses induced was also higher against RT and all its variants than in WT+AT-2 (but not in comparison against Gag and Nef pools) and was significantly correlated with low levels of viral load. CD4+ T cell count level was not related with this response. On the other hand, flow cytometry assays confirmed that the percentages of T cells that expressed CD57 (senescence) and CD38 (activation) were higher in non-responders, as well CD28 levels were substantially reduced. Conclusion: In summary, NL4-3/DRT virions stimulated T-cell specific immune responses. Our results indicated that it could be considered as a future candidate to be used as an effective immunogen or as an additional reagent for screening HIV-1specific responses in HIV seropositives and vaccines.

AIDS Vaccine 2011

149

POSTERS

Posters
P11.05

Topic 11: Novel Immunogens/Inserts P11.06


Generation of a Collection of HIV-1 Envelopes to Be Used as Immunogens with Increased Affinity for Broadly Neutralizing Antibodies
V. Snchez-Merino1, C.B. Ferreira1, A. Merino-Mansilla1, J.M. Gatell2, E. Yuste1 Institut dInvestigacions Biomdiques August Pi i Sunyer HIVACAT, Barcelona, Spain; 2HIVACAT, Hospital Clinic, Faculty of Medicine, University of Barcelona, Barcelona, Spain
1

Generation and Characterization of Retroviral Vectors Based on HIV-1 Genome as Novel Immunogens
C.C. Alvarez-Fernndez1, A.C. Guardo1, J.J. Garca-Perez2, F.F. Garca3, J.J. Gatell3, J.J. Alcam2, M.M. Plana1, S.S. Snchez-Palomino1 Fundacio Clinic, Barcelona, Spain; AIDS Immunopathology Unit, National Center of Microbiology, ICSIII, Madrid, Spain; 3Infectious Diseases Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
1 2

Background: The generation of new immunogens able to elicit strong immune-specific responses remains a major challenge to obtain a vaccine against HIV/AIDS. To this aim we have designed and produced a defective recombinant virus based on the HIV-1 genome that generates infective but non-replicative virions. Methods: Viral particles were generated through transient transfection in 293-T cells, of a full length HIV-1 DNA carrying a deletion of 892 aminoacid in the pol gene, encompassing the sequence for the reverse transcriptase (NL4-3/DRT). Wild-type viruses (NL4-3) inactivated with Aldritiol-2 or by treatment with a protease inhibitor (Amprenavir) were used as controls. Expression and processing of viral proteins were examined by Western-Blot. Infectivity and replication of viral particles were tested in PBMC using Gag-GFP labeled virions and in TZM-bl cells by luciferase activity. Viral particles so generated and characterized by cryoelectron microscopy were used to pulse lymphocytes from HIVinfected patients. The immunogenic capacity of these particles was assessed measuring interferon-G production by ELISPOT. Results: Defective particles were produced by NL4-3/DRT transient transfection and resulted, as expected, in infective but nonreplicative virions because of the absence of reverse transcriptase. Further analysis of purified virions by cryo-electron microscopy revealed an immature morphology characterized by an electrondense ring at the periphery and the absence of the canonical core in contrast to the AT-2-inactivated NL4-3 viruses. Preliminary in vitro assays showed that NL4-3/DRT was able to induce specific cellular immune responses and higher than AT-2-inactivated wild type HIV-1 viruses. Our results show that the lack of viral maturation may increase immunogenicity, which is supported by the enhancement of immunogenicity observed in immature virions generated by protease inhibitors treatment (Amprenavir). Conclusion: Immature HIV-1 virions generated from NL4-3/DRT represent effective novel immunogens displaying a safer and stronger capacity to induce HIV-specific cellular responses than wild-type viral particles.

Background: Antibody-based HIV-1 vaccine strategies rely on the elicitation of broadly neutralizing antibodies (bNAbs). Considering that the ability of an epitope to elicit antibodies will depend on its exposure on the virion, we took a new approach to optimize the HIV-1 envelope protein (Env) as an immunogen based on the selection of variants with increased affinity for the bNAb 4E10 from a library of virions with randomly mutated Env. Methods: We used the full-length env gene from HIV strain AC10 to generate the library of randomly mutated envelopes. Cloning was performed into NL4-3 context and virions were produced by transient transfection in 293T cells. Selection of viruses with increased affinity to 4E10 was carried out by an improved in-solution virion capture assay. RNA extraction and reverse transcription PCR of the env gene was performed from the virus population captured for further sequencing and cloning into NL4-3 context. Results: After a single round of selection we succeeded in selecting an envelope with a 4-fold increased affinity to 4E10 compared to AC10. Sequencing of the full-length envelope revealed 10 amino acid substitutions across the env gene, including the loss of 3 potential N-linked glycosylation sites. Mutation C131Y is especially relevant because it disrupts the architecture of the V1/V2 loop. Conclusion: We have selected, in a single round, an Env variant with increased affinity for 4E10 which harbours the kind of mutations that have been previously associated with increased neutralization sensitivity and increased induction of neutralizing antibodies. The immunogenicity of this variant will be tested next in a mouse model. We propose the usage of the present new approach for the selection of envelopes with increased affinity for different bNAbs and the generation of a collection of envelopes to be used as immunogens for eliciting a broadly neutralizing antibody response.

POSTERS 150 AIDS Vaccine 2011

Topic 11: Novel Immunogens/Inserts P11.07


A Prime-Boost Immunization with an Auxotrophic BCG Expressing HIV-1 Gag and Gag VLPs Induces Broad, Polyfunctional Memory T Cell Responses in Baboons
G.K. Chege1, W.A. Burgers1, A. Meyers1, H. Stutz1, A. Kiravu1, R. Bunjun1, R. Chapman1, E.G. Shephard1, E.P. Rybicki1, A. Williamson1
1

Posters

University of Cape Town, Cape Town, South Africa

Background: We previously reported a BCG pantothenate auxotroph expressing HIV-1 subtype C Gag (panBCG) that is immunogenic in baboons. In this study, we assessed the breadth, polyfunctionality and memory phenotype of T cell responses. Methods: Two groups of baboons (n=6 each) were primed or mock-primed twice with either panBCG or a control BCG and boosted with Gag VLPs. The breadth of Gag-specific responses was measured by IFN- ELISPOT method, using HIV-1 Gag peptides. The proportion of Gag-specific cells was measured by polychromatic flow cytometry after staining for IFN-, IL-2 and TNF-. CD28 and CD95 were used to delineate central and effector memory T cells. Results: PBMC from panBCG-primed animals responded to an average of 11 peptides per animal, targeting 3 peptides each in p17 and p24, and 5 peptides in p15 domains. In contrast, PBMC from controls targeted an average of 4 peptides per animal in either p24 or p15 domains. All panBCG-primed animals generated Gagspecific polyfunctional T cells, with the majority of responding CD4+ T cells producing all 3 cytokines simultaneously. The majority of CD8+ T cells produced IFN- and TNF- only. In contrast, only 2 of 6 controls mounted Gag-specific responses. In addition, panBCG-primed animals generated significantly higher frequencies of total cytokine positive T cells than the controls. Over 97% of the mean frequency of responses in the CD4 compartment was of a central memory phenotype. A more balanced central and effector memory Gag-specific CD8+ T cell phenotype was generated, with a mean of 69% and 31% in each compartment, respectively. Measurable CD4 and CD8 responses persisted in 2 of 6 and 1 of 6 animals respectively over a 12-week period after the last Gag VLPs inoculation. Conclusion: These data demonstrate that these candidate HIV vaccines are capable of inducing broad and polyfunctional T cells that may be long-lived.

AIDS Vaccine 2011

151

POSTERS

Posters
P12.01

Topic 12: Pediatric/Adolescent Infections and Trials P12.02


PedVacc Infant HIV-1 Vaccine Trials: First Stage Towards a Vaccine Against Breast Milk Transmission of HIV-1
T. Hanke1, K. Flanagan1, W. Jaoko1, G. John-Stewart1, M. Reilly1, B. Payne1, M. Afolabi1, G. Ambler1, A. Black1, N. Borthwick1, C. Gichuhi1, M. Mundia1, J. Ndure1, I. Njuguna1, J. Osanya1
1

HIV Neutralization in Breast Milk Mirrors that of Plasma and Is Mainly Mediated by IgG Antibodies
G. Fouda2, N. Yates2, J. Pollara2, G. Overman2, T. Mahlokozera1, A. Wilks1, H. Kang1, G. Ferrari2, G. Tomaras2, D. Montefiori2, S. Permar1
1

Harvard University, Boston, MA, USA; 2Duke University Medical Center, Durham, NC, USA

University of Oxford, Oxford, United Kingdom (Great Britain)

Background: Despite months of mucosal virus exposure, the majority of breastfed infants born to HIV-infected mothers do not become infected, raising the possibility that immune factors in milk inhibit mucosal transmission of HIV. HIV envelope -specific antibodies are present in the milk of HIV-infected mothers, but their functional role has not been fully investigated. Methods: The HIV envelope-specific antibody binding, heterologous virus neutralization and antibody-dependent cell cytotoxicity (ADCC) responses were measured in the breast milk and plasma samples of 40 HIV-infected, lactating women. In addition, the ability of breast milk and plasma to neutralize autologous breast milk and plasma viruses was determined in 7 women, including 3 who transmitted HIV to their baby during breastfeeding. Results: HIV envelope-specific IgG antibodies were more abundant than HIV-specific IgA in milk, although IgA is the predominant antibody isotype in milk. The concentration of anti-gp120 IgG was directly correlated in milk and plasma (r= 0.75 p<0.0001), yet the response in milk was two logarithms (logs) lower than in plasma. Autologous virus neutralization was detected in all plasma samples and in one of the transmitter breast milk. Heterologous virus neutralization and ADCC activity in milk were directly correlated to that in the systemic compartment, but were two logs lower in magnitude (neutralization r= 0.48, p=0.018, ADCC r=0.64 p<0.0001). Moreover, IgG purified from milk and plasma had equal heterologous neutralizing potency (r=0.65 p<0.0001), and the maximal ADCC potency was similar in the two compartments (r =0.60, p<0.0001). Milk heterologous virus neutralization titers also correlated with HIV gp120 envelopebinding IgG responses, but not IgA responses (r=0.70, p<0.0001 and r=0.13, p=0.34). No neutralization was detected in the nonIgG fraction of milk. Conclusion: These results suggest that plasma-derived IgG antibodies play a major role in the low level HIV neutralization and ADCC activity of breast milk.

Background: Over 60% of the global HIV-1-infected population lives in Africa and about half of the infected adults are women of childbearing age. Approximately half of mother-to-child transmission is due to breast-feeding, but formula feeding is not an option for many HIV-1 infected mothers. The best hope for protecting newborns and infants in developing countries against HIV-1 from their infected mothers while breastfeeding, is through development of a safe, effective, accessible vaccine. Methods: Two PedVacc infant HIV-1 vaccine trials examine the safety of a novel HIV-1 vaccine, MVA.HIVA, in infants. The trials are taking place in The Gambia and Kenya and will recruit in total 120 healthy, HIV-1-negative infants born to healthy, either HIV-1-positive or HIV-1-negative mothers. Both trials entail a single injection into the muscle of infants aged 20 weeks of MVA. HIVA. Any HIV-1-positive women in this study are provided with antiretroviral drugs and extensive feeding counselling during pregnancy and while breastfeeding to reduce the risk of HIV-1 transmission to their infants. Half of the infants in the study are randomised to receive the MVA.HIVA study vaccine in addition to their regular childhood immunizations. The other half only receive their regular immunizations, but not the study vaccine, and they serve as a control to the vaccinated infants. Results: Initial MVA.HIVA vaccine safety and immunogenicity data will be presented. Conclusion: The two PedVacc trials contribute to the capacity building for infant vaccine trials in Africa and represent the first stage towards a more complex heterologous prime-boost vaccine regimen.

POSTERS 152 AIDS Vaccine 2011

Topic 12: Pediatric/Adolescent Infections and Trials P12.03


Infant Feeding Strategy Is Not Associated with Breast Milk HIV-1 Viral Load
R.-. Rossenkhan1, V. Novitsky2, T.K. Sebunya3, L. Jean4, H.E. Jose5, S. Moyo5, L. Smeaton4, L. Shahin2, R. Musonda2, N. Thumbi5, S. Gaseitsiwe5, I. Thior5, M. Mmalane5, J. Makhema5, M. Essex2, R. Shapiro2
1 University of Botswana/ Botswana Harvard AIDS Institute Partnership, Boston, MA, USA; 2BHP/HSPH, Boston, MA, USA; 3 University of Botswana, Gaborone, Botswana; 4Harvard School of Public Health, Boston, MA, USA; 5Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana

Posters

Background: Exclusive breastfeeding has been associated with a reduced risk of late vertical HIV transmission as compared to an infant diet composed of breast milk mixed with supplemental foods or liquids. Hypothesized mechanisms include increased infectivity of breast milk from mothers who practice mixed breastfeeding (MBF), or mechanisms such as increased gastrointestinal permeability in the infant caused by mixed feeding. It has been proposed that MBF may result in subclinical mastitis and higher breast milk HIV titers. However, little is known about the relationship between feeding strategy and breast milk viral load. Methods: We therefore measured the HIV-1 concentration in breast milk in a sub-cohort of women enrolled in a mother to child HIV transmission prevention trial (the Mashi study). Infants born to mothers randomized to the breast feeding arm were categorized as exclusively breast-fed (EBF) or mixed-breastfed (MBF) based on information reported by the mother at birth and at subsequent monthly follow-up visits. Breast milk viral RNA was extracted using the High Pure System Viral Nucleic Acid Kit (Roche) for manual specimen preparation according to manufacturers instructions. Cell-free HIV-1 viral load was measured in breast milk samples using the COBAS TaqMan Analyzer (Roche Diagnostics Corporation, Indianapolis, USA). The lower limit of detection was 40 copies/ml HIV-1 RNA. Results: No significant differences in breast milk HIV-1 RNA levels were observed between EBF and MBF groups at either 2 weeks, 2 months, or 5 months postpartum (p=0.57, p=0.35, p=0.88, respectively) Conclusion: We report no observed relationship between MBF and measured breast milk viral RNA load.It appears unlikely that the increased transmission risk associated with MBF is mediated by breast milk HIV RNA concentration.

AIDS Vaccine 2011

153

POSTERS

Posters
P13.01

Topic 13: Preclinical and Clinical Therapeutic Trials

Safety and Immunogenicity of a HLA Supertype Restricted CTL Epitop Based HIV-1 Therapeutic Peptide Vaccine in CAF01 Adjuvant
I. Karlsson1, L. Brandt1, L. Vinner1, P. Andersen2, J. Gerstoft3, G. Kronborg4, A. Fomsgaard1 Statens Serum Institut, Copenhagen, Denmark; 2Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark; 3Department of Infectious Diseases, University Hospital Copenhagen, Copenhagen, Denmark; 4 Department of Infectious Diseases, University Hospital Hvidovre, Hvidovre, Denmark
1

Background: We designed a peptide based vaccine based on 18 peptides together with a novel adjuvant CAF01. CAF01 is a synthetic two-component liposomic adjuvant comprising the quaternary ammonium dimethyl-dioctadecyl-ammonium (DDA) and the immune modulator trehalose 6,6-dibehenate (TDB). The peptides were 15 CTL epitopes plus three T helper epitopes, in order to induce a balanced CD4+ and CD8+ cellular immune response. The selected CTL epitopes were derived from conserved regions of the HIV-1 and were subdominant (i.e. infrequently targeted) within natural infection. Moreover, the CTL epitopes were predicted to be restricted to five different HLA supertypes, thereby the vaccine should theoretically cover >90% of any population worldwide. Methods: In a single blinded phase I/II therapeutic HIV-1 vaccine trial 9 treatment nave HIV-1 infected Danish individuals where immunized intramuscular (week 0, 2, 4 and 8) with peptides (18 times 0.3 mg) together with the adjuvant CAF01 and 2 individuals received placebo (NaCl). Novel T cell responses were evaluated by IFN ELISpot two weeks and three months after the last immunization. Results: Previously undetected T cell responses specific for one or more epitopes were induced. The immunizations were safe and well tolerated. No allergic or autoimmune reactions were observed. No haematological, hepatic, muscular, pulmonary or renal toxicities were observed by blood testing. No overall or sustained change in viral load or CD4+ T cell counts was observed. Conclusion: In this study we show that it is possible to generate new T cell responses in treatment-nave HIV-1-infected individuals using peptides in adjuvant and thereby redirect immunity to target selected subdominant CTL epitopes. Although possible during untreated infection and high viral loads, therapeutic immunization during successive antiretroviral therapy where the immune functions are better preserved might lead to more potent and durable cellular responses.

POSTERS 154 AIDS Vaccine 2011

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.01


Long-Term Consequences of Participation in HIV Preventive Vaccine Trials Among Healthy Uninfected Subjects: The ANRS COHVAC Cohort
C. Durier1, C. Desaint2, B. Silbermann2, J. Lelivre3, B. Phung2, L. Slama4, P. Morineau-Le Houssine5, L. Cuzin6, I. Poizot-Martin7, P. Duchet-Niedziolka2, V. Dor8, V. Rieux8, A. Ross8, Y. Lvy3, G. Pialoux4, J. Aboulker1, B. Spire9, O. Launay2, On behalf of the ANRS COHVAC Study Group and the ANRS HIV Vaccine Network/VRI 8
1

Posters

P14.02
Multifunctional Fusion Proteins of the Human Engineered Antibody Domain m36.4 with Human Soluble CD4 and Fc as Potential Prophylactics
W. Chen2, X. Xiao2, C. Zhang1, Z. Zhu2, Y. Wang2, H. Goldstein1, D.S. Dimitrov2
1 2

Albert Einstein College of Medicine, New York, NY, USA; National Cancer Institute-Frederick, National Institutes of Health, Frederick, MD, USA

INSERM SC10, Villejuif, France; 2Universit Paris-Descartes; INSERM CIC BT 505; AP-HP, Hpital Cochin, Paris, France; 3 Hpital Henri Mondor, Universit Paris-Est Crteil, INSERM U955, Crteil, France; 4AP-HP, Hpital Tenon, Paris, France; 5 CHU Hpital Htel-Dieu, Nantes, France; 6Hpital Purpan, Toulouse, France; 7CHU Sainte-Marguerite, ImmunoHmatologie Clinique, Marseille, France; 8ANRS, Paris, France; 9 INSERM U912, IRD, Universit de la Mditerrane - AixMarseille II, Marseille, France
Background: ANRS COHVAC cohort gathers data of healthy low HIV risk volunteers who participated in phase I and II trials in France since 1992; one of its objectives is to explore long term psycho-social consequences of participation in HIV vaccine trials and vaccine-induced seropositivity (VISP). Methods: Vaccine candidates were a recombinant envelope protein (rgp160) [1992-1993], Alvac vectors (vCP) expressing Env, Gag, Pro and CTL domains of Pol [1994-2001], and/or HIV-1 lipopeptides representing CTL epitopes of Gag, Pol and Nef proteins [1997-2007]. Personal and social consequences of participation were collected by an anonymous questionnaire and an interview with a clinician. VISP was defined as HIV positivity in at least one licensed enzyme immunoassay, regardless of Western-blots results. Results: 422 volunteers were eligible for the cohort study. Questionnaires and blood samples were collected from 250 (59%) and 248 subjects at inclusion, respectively. The median age was 52 years (25-71), 52% were male. Most subjects had graduated from high school (86%), were employed (84%) and 102 (40%) were blood or tissue donors. VISP was observed in 11% (27/248) of subjects: 66% (19/29), 16% (7/44) and 0.6% (1/151) of rgp160, vCP and lipopeptides recipients, respectively. Western-blots showed presence of bands included in the vaccine products (mainly gp160, p25, p55) in 10 of these subjects. The median VISP durability was 16.6 years (range, 15.6 to 18.3) for rgp160 and 9.7 years (range, 8.5 to 15) for vCP. Five subjects reported problems with insurance and 32 with blood or tissue donation after participation had stopped. Other social negative consequences were expressed by 13% of interviewed subjects. Conclusion: Negative consequences were seldom reported except regarding blood donation. VISP was maintained 16 years in a majority of rgp160 recipients and over 8 years in a minority of Alvac recipients. Volunteers should be informed of possible VISP several years after active participation.

Background: Monoclonal antibody-based prophylactics are safe and highly efficacious in preventing disease by passive immunization with the prominent example of Synagis against the respiratory syncytia virus. However, HIV-1 is highly variable and quickly develops resistance to antibody-based inhibitors. We have hypothesized that targeting two highly conserved receptor sites on the HIV-1 envelope glycoprotein (Env), the CD4 binding site (CD4bs) and the coreceptor binding site (Cobs), could allow broad and potent protection. To test this hypothesis we proposed to join highly potent antibodies against the Cobs with several variants of soluble CD4 (sCD4) and IgG1 Fc, and test them against various HIV-1 isolates in vitro and in vivo. Methods: A previously identified engineered antibody domain (eAd) m36 (Chen et al., PNAS 2008) was further improved by panning and screening large m36 mutant libraries against Envs and Env-sCD4 complexes. Multifunctional fusion proteins were constructed by using the improved version of m36, m36.4 (targeting the Cobs), sCD4 (targeting the CD4bs), and Fc ensuring long half-life in circulation. They were tested for binding to the Env and inhibitory activity against isolates from different clades in standard assays, and protection of humanized NOD/SCID/ cnull mice against HIV-1 infection. Results: The m36.4-sCD4-Fc fusion proteins bound to recombinant Envs with high avidity in ELISA and exhibited exceptionally high potency against all tested isolates in a pseudovirus neutralization assay, significantly higher than that of m36.4 or sCD4 alone. They protected 4 of 6 humanized NOD/SCID/cnull mice from infection by HIV-1 isolate JRCSF. Conclusion: Our data suggest that m36.4 derivatives are promising HIV-1 candidate prophylactics and tools to study highly conserved gp120 structures with implications for understanding mechanisms of entry and design of vaccine immunogens.

AIDS Vaccine 2011

155

POSTERS

Posters
P14.03

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.04


HIV-Specific CD4 and CD8 Proliferative Responses and Effector Memory Profile in Swedish Vaccinees Immunized with HIV-1 DNA and HIV-1 MVA and HIV-1 MVA
K. Godoy-Ramirez1, C. Nilsson2, L. Jernberg1, K. Karln1, M. Marovich3, B. Hejdeman4, B. Wahren 2, E. Sandstrm4, G. Biberfeld2
1

Safety and Reactogenicity of ALVAC-HIV (vCP1521) and HIV-1 gp120 AIDSVAX B/E PrimeBoost Vaccination Regimen in a Community-Based Efficacy Trial
P. Pitisuttithum1, S. Rerks-Ngarm2, V. Bussaratid1, J. Dhitavat1, W. Maekanantawat1, S. Pungpak1, P. Suntharasamai1, S. Vanijanonta1, S. Nitayapan3, J. Kaewkungwal1, S. Chunsuttiwat2, N. Premsri2, M. Benenson3, P. Morgan3, J. Berenberg4, S. Gurunathan5, D.P. Francis6, J. Chiu3, J. Excler7, M.L. Robb7, D. Stablein8, N.L. Michael7, J. Kim7 Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; 2Department of Disease Control, Ministry of Public Health, Bangkok, Thailand; 3Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 4Department of Medicine, Tripler Army Medical Center, Honolulu, Hawaii, USA; 5sanofi pasteur, Swiftwater, PA, USA; 6Global Solutions for Infectious Diseases, South San Francisco, CA, USA; 7U.S.Military HIV Research Program,Walter Reed Army Institute of Research, Rockville, MD, USA; 8EMMES Corporation, Rockville, MD, USA
1

Swedish Institute for Communicable Disease Control, Stockholm, Sweden; 2Swedish Institute for Communicable Disease Control and Karolinska Institute, Stockholm, Sweden; 3US Military HIV Research Program, Walter Reed Army Institute of Resear, Rockville, MD, USA; 4Venhlsan, Karolinska Institutet, Sdersjukhuset, Stockholm, Sweden
Background: We used a Flow-Cytometric Lymphoproliferation Assay (FC-LPA) to monitor HIV-1-vaccine-induced responses. Healthy volunteers were immunized at 0, 1 and 3 months with DNA plasmid expressing gp160 of HIV-1 subtypes A, B and C; revB; p17/p24 gag A and B and RTmut B. At nine months they were boosted with a heterologous MVA expressing HIV-1 env, gag, pol of CRF01_AE. Approximately 3 years after the first HIVMVA vaccination, 24 volunteers were given a second HIV-MVA boost and the last 17 vaccinees were tested by FC-LPA. Methods: Lymphoproliferative responses to AT-2 inactivated HIV-1 antigen (donated by Jeffrey Lifson, NCI, USA), were monitored by FC-LPA two weeks after the second HIV-MVA vaccination. Fresh cells were incubated with or without HIV-1 antigen for 6 days and stained with a T-cell panel (CD3/CD4/ CD8). The read-out was based on identification of lymphoblasts (%) and the result presented as stimulation index (SI). Twelve of the samples were simultaneously stained for memory markers (CD45RA/CCR7/CD28/CD27). Results: All of 17 tested samples (100%) were reactive in the CD4 T-cell compartment and 14/17 (82%) in the CD8 T-cell compartment. The CD4 T-cell blasts were predominantly of effector memory type (CD45RA-CCR7-CD28+CD27-) but approximately a tenth showed a central memory profile (CD45RA-CCR7+CD28+CD27-). The CD8 T-cells blasts were also predominantly of effector memory type but several stages of differentiation were observed. The proportion of central memory cells was approximately 20%. Conclusion: HIV-specific T-lymphocyte proliferative responses were detected in all volunteers immunized with HIV-DNA followed by HIV-MVA. The FC-LPA revealed both CD4+ and CD8+ T-cell proliferation with a predominant effector memory phenotype. A sensitive and robust FC-LPA, which allows immunophenotyping of the immune responses may be an alternative to the conventional 3H- thymidine uptake assay for assessment of vaccine-induced T-cell proliferation.

Background: A prime-boost vaccination regimen with ALVACHIV (vCP1521) administered intramuscularly at 0, 4, 12, and 24 weeks and gp120 AIDSVAX B/E at 12 and 24 weeks demonstrated modest efficacy of 31.2% for HIV acquisition in HIV-uninfected adults in a phase III trial in Thailand. Methods: Reactogenicity was recorded for 3 days following vaccination. Adverse events were monitored every 6 months for 3.5 years, and pregnancy outcomes were recorded. Results: Of the 16,402 volunteers, 69% of the participants reported an adverse event any time after the first dose. Only 32.9% of events occurred within 30 days post vaccination, 3.2% attributed to vaccine, and rates did not differ between groups. The frequency of serious adverse events was similar in vaccine (14.3%) and placebo (14.9%) recipients (p=0.33). None of the 160 deaths (85 in vaccines, 75 in placebo) were related to vaccine. The most common cause of death was trauma and accident. Approximately 30% of female participants reported a pregnancy during the study. Abnormal outcomes were experienced in 17.1% and 14.6% (p=0.13) of vaccine and placebo recipients with pregnancies, respectively. Among pregnancies with estimated dates of conception within 3 months of a vaccination, the large majority of these abnormal outcomes were spontaneous or elective induced abortions reported in 22.2% and 15.3% of vaccine and placebo pregnant recipients, respectively (p=0.08). Local reactions (mostly pain and tenderness) occurred in 88.0% of vaccine and 61.0% of placebo recipients (p<0.001) and were more frequent after ALVAC-HIV than AIDSVAX B/E vaccination. Systemic reactions (mostly fatigue, myalgia, headache) were more frequent in vaccine than placebo recipients (77.2% vs. 59.8%, p<0.001). Local and systemic reactions were mostly mild to moderate, resolving within 3 days.

POSTERS

Conclusion: The ALVAC-HIV and AIDSVAX B/E vaccine regimen was found to be safe, well tolerated and suitable for large-scale use in Thailand.

156

AIDS Vaccine 2011

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.05


Construction and Characterization of HIV-1 Mosaic Modified Vaccinia Ankara (MVA) Vaccines
L. Eller1, J. Currier1, S. Ratto-Kim1, M. Robb1, J. Kim1, M. Eller1, D. Barouch2, B. Moss3, N. Michael1, M. Marovich1, P. Earl4 US Military HIV Research Program, Rockville, MD, USA; Division of Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; 3Laboratory of Viral Diseases, National Institute of Allergy and I, NIH, Bethesda, MD, USA; 4 Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD, USA
1 2

Posters

P14.06
Persistence of Vaccine-Induced Antibodies Following HIV-1 DNA Prime MVA Boost Vaccination Among Healthy Tanzanian Volunteers
S. Aboud1, P. Munseri2, A. Joachim3, M. Bakari3, C. Nilsson3, D. Buma4, E.A. Aris4, L.F. Eligius3, B. Andreas5, M. Robb6, M. Marovich6, N. Michael6, B. Wahren5, G. Biberfeld5, F. Mhalu3, E. Sandstrom7
1

Background: Several Modified Vaccinia Ankara (MVA) vaccines containing natural HIV-1 sequences are currently showing promise in clinical trials. Recently, polyvalent HIV-1 mosaic immunogens have demonstrated increased breadth and depth of cellular immune responses in primates. Here, we describe construction and preliminary characterization of HIV-1 mosaic MVA vaccines. Methods: Two double recombinant MVA viruses were constructed, each containing the previously described M1 and M2 mosaic env, gag, and pol sequences, via successive rounds of plaque purification and amplification. Balb/c mice were immunized with 10^7 pfu of Mosaic MVA viruses at 0 and 4 weeks. Intracellular cytokine staining (ICS) and pentamer staining using KETI and KDTI gag peptides were performed 1 week after the 2nd immunization. Binding antibody titers to subtype A/E, C, D and A gp140 envelopes were evaluated 2 weeks after the 2nd immunization by ELISA. Results: Two MVA viruses containing mosaic HIV-1 env/gag/ pol inserts were constructed and expression of proteins was confirmed by immunostaining and Western blot. Stability of viruses was confirmed by PCR, sequencing and immunostaining after 10 rounds of serial passage. Mosaic MVAs generated positive CD8 T cell responses in mice to KETI and KDTI gag peptides by pentamer staining and also by production of IFN-gamma by ICS. Immunogenicity was also demonstrated by pooled peptide ELISPOT responses in mice. Binding antibodies to envelopes of diverse subtypes were generated at titers comparable to previously constructed MVA vaccines. Conclusion: We demonstrate that HIV-1 mosaic MVA vaccines are comparable to previously constructed MVA vaccines in terms of titer, stability, expression and immunogenicity in mice. HIV-1 Mosaic MVA vaccines, in combination with HIV-1 Ad26 Mosaic vaccines, are currently undergoing evaluation in non human primate studies where evaluations of breadth and depth of the cellular immune response will be fully characterized.

Muhimbili University College of Health Sciences, Dar es Salaam, United Republic of Tanzania; 2Venhlsan, Karolinska Institutet, Sdersjukhuset, Stockholm, Sweden; 3Muhimbili University of Health and Allied Sciences, Dar es Salaam, United Republic of Tanzania; 4Muhimbili National Hospital, Dar es Salaam, United Republic of Tanzania; 5Swedish Institute for Communicable Disease Control, Solna, Sweden; 6Walter Reed Army Institute of Research, Rockville, MD, USA; 7Venhalsan, Karolinska Institutet, Sodersjukhuset, Stockholm, Sweden

Background: A phase I/II HIV vaccine trial (HIVIS03) using a multiclade, multigene HIV-1 DNA prime with two heterologous HIV-1 MVA boosts has been completed among healthy adults in Dar es Salaam, Tanzania. Methods: Sixty HIV-uninfected volunteers randomized to three groups of 20, received DNA plasmids expressing HIV-1 gp160 subtypes A,B,C;revB;p17/p24gagA,B and Rtmut at 1 mg intradermally or 3.8 mg intramuscularly or placebo at months 0, 1 and 3 using a needle-free injection device (Biojector). Volunteers were boosted intramuscularly with 108 plaque-forming units of recombinant MVA expressing HIV-1 env, gag, pol of CRF01_AE or placebo at months 9 and 21. The volunteers were followed up 17-22 months after the second HIV-MVA boost and the presence of vaccine-induced antibodies was analyzed. Diagnostic HIV serological testing was performed using Murex (Abbott Murex, UK) and Integral (Siemens, Germany) HIV antigen/antibody ELISAs and Inno-Lia immunoblot (Inno-genetics, Belgium) assays. Results: None of the vaccinees or placebo recipients was positive in the diagnostic HIV serological assays after the three HIV-DNA immunizations or after the first HIV-MVA boost. Four weeks after the second HIV-MVA boost, all 30 vaccinees (100%) were positive in all three diagnostic HIV assays. On the immunoblot assay, all 30 showed reactivity to both Gag (p24) and Env (gp120 and/or gp41). Twenty-nine out of 30 (97%) vaccinees were still reactive in both Murex and Integral HIV antigen/antibody ELISAs 17 to 22 months after the second HIV-MVA boost. On the Immunoblot assay, 21 out of 30 (70%) vaccinees fulfilled the diagnostic criteria for seropositivity and 9 were indeterminate. All of 30 (100%) vaccinees reacted against Gag and 21 out of 30 (70%) reacted against Env (gp120 or gp41). Testing by the Roche HIV-1 DNA PCR assay excluded HIV-1 infection in all these volunteers. Conclusion: This study showed the durability of HIV-DNA/MVA vaccine-induced antibody responses in Tanzanian volunteers.

AIDS Vaccine 2011

157

POSTERS

Posters
P14.07

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.08


Lipopeptide Based HIV Vaccine Efficiently Primes Recombinant MVA (HIV-Clade B Gag/Pol/Nef) Elicited HIV-Specific T Cell Responses
N. Dereuddre-Bosquet2, I. Mangeot-Mderl2, V. Contreras2, A. Cosma2, M. Baron2, F. Martinon2, Y. Lvy1, R. LeGrand2
1

Characterization of ANRS HIV LIPO-5 Vaccine in Healthy Volunteers Combining Cytokine Multiplex and Transcriptomic Analyses
S. Hue1, L. Richert4, H. Hocini1, M. Surenaud1, P. Tisserand1, C. Lacabaratz1, D. Salmon-Cron2, M. Raimbault4, J. Lelivre3, B. Liquet4, Y. Levy1, R. Thiebaut4 Universit Paris Est, Crteil - Hpital Henri Mondor, Crteil, France; 2Universit Paris Descartes - Hpital Cochin, Paris, France; 3Universit Paris Est, Crtail - Hpital Henri Mondor, Crteil, France; 4INSERM U897 - the ANRS HIV Vaccine Network/Vaccine Research Institute, Bordeaux, France
1

INSERM U955, Facult de Mdecine Crteil and ANRS HIV Vaccine Network, Crteil, France; 2CEA and UMR E1, Universit Paris-Sud, Fontenay-aux-Roses, France

Background: ANRS Vac 18 study evaluated the immunogenicity of HIV-LIPO-5 vaccine (Gag17-35, 253-284, Pol325-355, Nef6697 and 116-145, coupled to a palmytoil tail) administered at weeks 0, 4 and 12 in healthy volunteers. 62-69% of vaccinees developed HIV-specific CD8 (12-days PBMC cultured IFN- Elispot using optimal peptides). Here, we measured cytokine and gene expression profiles associated with vaccine responses. Methods: PBMCs from 12 vaccinees (W0 and 14) were stimulated with either LIPO-5 vaccine, or a pool of 15-mers Gag peptides. Transcriptomic (Illumina) and cytokine multiplex (Millipore) analyses were performed at 6/24H and D11, respectively. Statistical analyses (Wilcoxon for paired samples) were performed on concentration and raw fluorescence. Normalization and filtering were performed to select detected transcripts, and various conditions were compared using a false discovery rate set at 0.1. Results: Gag Specific responses detected at W14 as compared to W0 were characterized by an increase of IFN-g, TNF-a, IL-5, IL-10, IL-13 cytokines (all P values <.01). An adjuvant effect of Lipopeptide tail was characterized at W0 by an induction of IFN-g and IL-10 in the supernatants of PBMC stimulated with LIPO5. Consistently, a significant variation of gene transcripts after 6 hours (n=160 probes) and 24 hours (n=8 probes) stimulation was observed in LIPO-5 stimulation at W0. However, following vaccination, 3305 and 1821 probes changed significantly after 6 and 24 hours of LIPO-5 stimulation, respectively. Among these genes, IFN-g, CXCL9, CXCL10, IL2RA, TNFAIP6, CCL3L1, IL-6 increased significantly (fold change >2). Conclusion: Specific responses to LIPO-5 vaccination are characterized by Th1 and Th2 profiles. The signature profile of LIPO-5 stimulation before vaccination could provide information about the adjuvant effect of lipid tail. Consistently with cytokine responses, vaccination is specifically associated with a large number of gene expression. This approach might help to identify new signatures associated with HIV vaccine responses.

Background: HIV-lipopeptide candidate vaccine (Lipo-5) developed by the ANRS has been shown to induce HIV specific precursor T cells in more than 70% of healthy volunteers. The objective was to assess, in nonhuman primates (NHP), rMVAHIV/lipo-5 and Lipo-5/rMVA-HIV prime/boost combinations. Methods: Five groups of four cynomolgus macaques were injected 1) group 1 (L/L): with Lipo-5 alone (250 g by the intramuscular route) on weeks 0, 8, 16 and 21, 2) group 2 (rM/L) with Lipo-5 (weeks 16 and 21) after a prime (weeks 0 and 8) with the ANRS rMVA (5x107 pfu by the subcutaneous route) 3) group 3 (L/rM): with rMVA (weeks 16 and 21) after priming with Lipo-5 (weeks 0 and 8). Groups 4 and 5 are control groups with wt MVA. Lipo5 vaccine contains Gag17-35, Gag253-284, Pol325-355, Nef6697 and Nef116-145. Immune response was assessed using PBMC stimulated ex vivo with pools of Gag, Pol and Nef peptides and by analyzing the frequency of IFN-g and IL-2 producing cells by ELISpots, cytokine secretion in culture supernatants and by intracellular staining (ICS). Results: In L/L group no ELISpot IFN-g response was detectable. In rM/L maximal T cell responses were elicited in 4/4 NHP after two rMVA primes (mean+/-SD: 599 +/- 204 SFC/106 cells). In the group L/rM 0/4 NHP responded after the prime and 4/4 after a single injection of rMVA (mean+/-SD: 671+/-329 SFC/106 cells). In control groups wt MVA did not elicit any HIV specific response. Conclusion: Sequence of prime/boost combining ANRS Lipo5 and rMVA differently impacts HIV specific T cell responses. Lipo-5 primes efficiently for vector-induced HIV specific responses. This study provides a rationale for future clinical trials in human volunteers.

POSTERS 158 AIDS Vaccine 2011

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.09


Phase I Trial of a Prophylactic HIV-1 DNA gagpol-env-nef Vaccine in St. Petersburg, Russia
B. Murashev1, O. Nazarenko1, E. Akulova1, S. Verevochkin1, M. Astanina1, T. Krasnoselskih2, D. Lioznov2, E. Sokolovskiy2, A. Kozlov3 State Research Institute of Highly Pure Biopreparations, St. Petersburg, Russian Federation; 2St Petersburg Pavlov State Medical University, St. Petersburg, Russian Federation; 3The Biomedical Center, St. Petersburg, Russian Federation
1

Posters

P14.10
The HIV Vaccine Candidate MVA-B Triggers Robust, Polyfunctional and Memory T Cell Responses to HIV-1 Antigens in a Phase I Clinical Trial in Spain
C. Gmez1, J. Njera1, B. Perdiguero1, J. Garca-Arriaza1, C.S. Sorzano1, V. Jimnez1, R. Gonzlez-Sanz1, J. Jimnez2, M. Muoz-Fernndez2, J. Lpez Bernaldo de Quirs2, A.C. Guardo3, F. Garca3, J.M. Gatell3, M. Plana3, M. Esteban1
1

Background: A phase I trial was conducted to assess the safety and immunogenicity of an HIV-1clade A FSU DNA vaccine in healthy HIV-1 uninfected adults. Recombinant DNA vaccine candidate (DNA-4) consists of four plasmids encoding consensus sequences of the nef, gag, pol (rt) and env (gp140) HIV-1 subtype A FSU genes. Methods: 21 participants were randomly divided into three groups (7 per group). Each group received 0.25, 0.5 or 1.0 mg of DNA-4, respectively. Vaccine was administrated four times intramuscularly at days 0, 6, 10 and 14. Immunogenicity was assessed by ex vivo IFN- ELISpot, IFN-/IL-2/TNF ICS, CFSE LPA and ELISA at days 14,25,43 and 60 after the first vaccination. The samples, which were taken before immunization, served as negative controls. Results: The vaccine regimen was found to be safe and well tolerated in this sample. 4 vaccinees from the first group (57%), 4 - from the second group (57%), and 2 from the third group (29%) were IFN- ELISpot reactive. In the ICS assays 7/7 (100%) subjects, who received 0.25 mg of DNA-4, 2/7 (29%) subjects, who received 0.5 mg of DNA-4, and 6/7 (86%) subjects, who received 1 mg of DNA-4, had CD3+CD8+ T-cell responses. CD3+CD8T-cell responses were detected in 6/7 (86%), 5/7 (71%), 4/7 (57%) subjects from first, second and third groups respectively. 4/7 (57%), 4/6 (67%), 3/7 (43%) patients had positive CD8+ LPA responses; and 5/7 (71%), 5/6 (83%), 3/7 (43%) demonstrated CD8- LPA responses. Altogether 17/21 (81%) subjects revealed CD8+ responses, and 19/21 (90%) CD8- responses. 5 of 21 vaccines exhibited vaccine-induced seropositivity, 3 to Nef, 1 to Gag, and 1 to gp140. Conclusion: DNA-4 vaccine is well tolerated. Vaccination with DNA-4 regimen elicited moderate CD8- and CD8+ T-cell responses, but modest humoral responses. This regimen should be considered as part of future vaccine strategies.

Centro Nacional de Biotecnologa, Madrid, Spain; 2Hospital Gregorio Maran, Madrid, Spain; 3Hospital Clinic-IDIBAPS, Barcelona, Spain

Background: There is general consensus that for an HIV/AIDS vaccine to be effective it must trigger broad, polyfunctional and durable antigen-specific B and T cell immune responses. Attenuated poxvirus vectors expressing HIV-1 antigens are considered promising HIV/AIDS vaccine candidates. However, the modest efficacy observed in the RV144 trial point to the development of improved pox vectors. Methods: Here we described the T cell immune responses induced in healthy volunteers participating in a phase I clinical trial in Spain (RISVAC02) after intramuscular administration of three doses of the recombinant MVA-B expressing monomeric gp120 and the fused Gag-Pol-Nef (GPN) polyprotein of clade B. Results: The majority (92.3%) of the volunteers immunized had a positive specific T cell response at any time post-vaccination as detected by IFN- ICS assay. The CD4+ T cell responses were predominantly Env directed, whereas the CD8+ T cell responses were similarly distributed against Env, Gag and GPN. The proportion of responders after two doses of MVA-B was similar to that obtained after the third dose of MVA-B vaccination and the responses were sustained (84.6% at week 48). Vaccine-induced CD8+ T cells were polyfunctional and mainly distributed within the TEM and TEMRA cell populations. Anti-vector T cell responses were mostly induced by CD8+ T cells, highly polyfunctional and of TEMRA phenotype. Conclusion: These findings demonstrate that the poxvirus MVA-B vaccine candidate given alone is highly immunogenic, inducing broad, polyfunctional and long-lasting CD4 and CD8 T cell responses. Thus, MVA-B can be considered as promising candidate HIV/AIDS vaccine vector for further clinical trials.

AIDS Vaccine 2011

159

POSTERS

Posters
P14.11

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.12 LB


Safety and Immunogenicity of DNA and MVA HIV-1 Subtype C Vaccine Prime-Boost Regimens: A Phase I Trial in India
S.M. Mehendale1, M. Thakar2, M. M 3, S. Sahay2, S. P3, A. Verma2, S. Kurle2, A. Shete2, S. Kochhar4, J. Gilmour5, R. Goyal5, L. Dally6, J.H. Cox5, J. Excler5, P. Fast5, K. V3, R. Paranjape2, R. VD3
1

Biological Correlation of HIV Exposure with Self-Reported High Risk Sexual Behavior Among Women
C.D. Zorrilla1, S.A. Abdulhaqq2, V.M. Tamayo-Agrait1, I. Febo-Vazquez1, S. Garced1, P. Piovanetti1, C. Tomescu2, L. Azzoni2, L.J. Montaner2
1 UPR School of Medicine, San Juan, PR, USA; 2The Wistar Institute, Philadelphia, PA, USA

Background: The recruitment of women with demonstrated high risk behavior for participation on preventive vaccine trials have been a difficult task. Among women enrolled in the STEP study there were not enough sero-conversions to carry out gender based analysis raising the concern that risk behavior had been overestimated Methods: To determine presence of HIV-1 exposure, cell-mediated responses against gag antigen were measured by flow-cytometry in women from 4 risk behavior groups: discordant couples (DC), commercial sex workers with high and moderate risk behavior (CSW-H; CSW-L), and monogamous unexposed controls (CG). In order to correlate sexual practices with a biological marker of previous exposure to HIV, an analysis between cohort behavior and presence of HIV-gag specific responses was performed. Samples from 58 of 79 women for whom HIV-gag specific responses were measured were used (intracellular IFN-; mean response= 0.45% of total CD8+ T-Cells expressing IFN-+). Results: Higher frequency of HIV-gag responses were seen in the sex workers than in the other 2 cohorts (CSW 85.2%; DC 7.4%; CG 7.4%; p<0.00001). Responses to HIV-gag were also higher when compared by number of sexual partners (<10 vs. >10) in the previous year (34.6% vs. 65.4% p= 0.026) or (<3 vs. >3) partners (19.2% vs. 80.8% p<0.00001). Higher frequency of responses were seen on women who reported cigarette smoking (90.5% vs. 9.5%, p<0.0001), had evidence of an STD (85.2% vs. 14.8%, p<0.0001), reported any drug use (63.0% vs. 37.0%, p=0.05) or injection drug use (92.6% vs. 7.4% p<0.0001). The analysis by lifetime sexual partners showed NS. Conclusion: HIV-gag responses as an indication of HIV-1 exposure in women who self-reported high risk behaviors were related to numbers of sexual partners in the past year, STDs and drug use. HIV-1 exposure in women with high risk behavior confirms that this population should be included into prevention trials.

National Institute of Epidemiology, Chennai, India; 2National AIDS Research Institute, Pune, India; 3Tuberculosis Research Center, Chennai, India; 4Institute for Oneworld Health, New Delhi, India; 5International AIDS Vaccine Initiative, London, United Kingdom (Great Britain); 6Emmes Corporation, Rockeville, MD, USA
Background: With over 2.5 million HIV-infected people and evidence of ongoing transmission, the search for a preventive HIV vaccine for Indian people gains importance. We have earlier reported moderate immunogenicity of MVA vaccine. Evidence elsewhere indicates superior immunogenicity of heterologous prime-boost strategy. We assessed the safety and immunogenicity of HIV-1 subtype C based DNA and MVA prime-boost regimens in Indian adults. Methods: Sixteen healthy HIV-uninfected volunteers (12 vaccine, 4 placebo) were randomly assigned to vaccine or placebo in group A (DNA at 0, 1 and MVA at 3, 6 months) or group B (MVA at 0, 1, 6 months). All vaccines were administered intramuscularly in the deltoid muscle. Reactogenicity was assessed at 3, 7 and 14 days post-vaccination; adverse events up to 9 months and serious adverse events through out the trial. T-cell and antibody responses were assessed pre and 1 and 2 weeks post each vaccination, then at 9, 12 and 18 months. Results: Local and systemic reactogenicity profiles were comparable between groups, and were mostly mild and transient. No serious adverse events were reported. In group A vaccine recipients IFN-gamma ELISPOT response was detected in 0%, 25%, 100% and 100% participants post 1st, 2nd, 3rd and 4th vaccinations and in 60%, 63.6% and 91.7% group B participants post 1st, 2nd and 3rd vaccinations, respectively. Responses were directed to multiple HIV proteins (magnitude of 108 250 SFU per 106 PBMC) in most volunteers. HIV-specific antibodies were detected in 10/12 and 11/12 vaccine recipients in groups A and B, respectively, and neutralizing antibodies to tier-1 viruses were detected against HIV SF162 and MW-965 in most individuals. Conclusion: Both vaccination regimens were found to be safe and well-tolerated. Heterologous DNA-MVA prime boost strategy elicited comparable T-cell immune responses to the homologous MVA strategy.

POSTERS 160 AIDS Vaccine 2011

Topic 14: Preclinical and Clinical Prophylactic Vaccine Trials P14.13 LB


Evaluation of Peripheral Blood and Mucosal HIV1 Viral Load in Volunteers Who Became HIV-1 Infected During Participation in the Thai Phase III Trial
S. Rerks-Ngarm1, M.S. de Souza2, J. Bowornwatanuwong3, N. Premsri1, V. Ngauy2, S. Li4, J. Kaewkungkal5, R. Trichavaroj6, N. Churikanont6, D. Francis7, S. Gurunathan8, J. Tartaglia8, R. OConnell9, R.M. Paris9, S. Nitayaphan10, N.L. Michael9, M.L. Robb9, P. Thongcharoen11, P. Kunasol1, P. Gilbert4, J.H. Kim9 Ministry of Public Health, Bangkok, Thailand; 2US Military HIV Research Program/Dept. of Retrovirology, AFRIMS, Bangkok, Thailand; 3Chonburi Regional Hospital, Chonburi, Thailand; 4SCHARP/Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 5Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; 6Dept. of Retrovirology, AFRIMS, Bangkok, Thailand; 7Global Solutions in Infectious Diseases, San Francisco, CA, USA; 8sanofi pasteur, Swiftwater, PA, USA; 9US Military HIV Research Program, Rockville, MD, USA; 10Royal Thai Army, Dept of Retrovirology, AFRIMS, Bangkok, Thailand; 11Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
1

Posters

P14.14 LB
Penile SIV Challenge of Ad5-Infected Macaques Immunized With an Ad5-Based Vaccine Recapitulates the Enhanced Infection Rate Among STEP Trial Vaccines
H. Qureshi5, Z. Ma5, Y. Huang1, G. Hodge5, M. Thomas2, J. DiPasquale2, V. DeSilva5, L. Fritts5, A.J. Bett3, J. Shiver3, M. Robert-Guroff2, M. Robertson3, D. Casimiro3, M. McChesney5, P. Gilbert4, C. Miller5
1

Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchins, Seattle, WA, USA; 2Vaccine Branch, NCI, NIH, Bethesda, MD, USA; 3Merck Research Laboratories, West Point, PA, USA; 4Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchin, Seattle, WA, USA; 5UC Davis, Davis, CA, USA
Background: The MRKAd5 HIV-1 subtype B Gag-Pol-Nef vaccine enhanced susceptibility to HIV-1 infection in Ad5-seropositive, uncircumcised men. Methods: To develop an animal model to understand these results we determined if rhesus macaques chronically infected with a host range mutant adenovirus type-5 (Ad5hr) and then immunized with a replication defective Ad5 SIVmac239 Gag-PolNef vaccine were more resistant or susceptible to SIV infection than unimmunized rhesus macaques during a series of repeated penile SIVmac251 exposures. Results: We found that 2 of 9 rhesus macaques infected with Ad5hr prior to immunization became SIV infected after penile exposure to 103 TCID50 of SIVmac251. In contrast none of the 34 unimmunized animals, including 8 Ad5 seropositive animals immunized with the empty vector; Ad5 seronegative animals immunized with the Ad5 SIV vaccine (n= 9) or empty vector (n= 9) and 8 nave control animals became SIV infected after penile exposure to the same dose of SIVmac251. At the lowest SIV exposure dose (103 TCID50), the risk of infection was greater for animals with Ad5 titers at the time they were immunized with the MRKAd5 SIVmac239 subtype Gag-Pol-Nef vaccine compared to all the other animal groups (p= 0.004 based on log rank test). Penile exposure to more concentrated virus inoculums produced similar rates of infection in all animals groups. The Ad5 SIV vaccine induced CD8+T cell responses in approximately 70% monkeys, which is similar to the proportion of humans that responded to HIV-1 vaccine. Conclusion: The results of the NHP study described here recapitulate the results of the STEP trial and demonstrate that the results of NHP studies using challenge viruses and routes of exposure designed to mimic the variety and complexity of HIV1 sexual transmission can reflect the outcomes of AIDS vaccine efficacy trials.

Background: The Thai Phase III trial of ALVAC-HIV and AIDSVAX B/E was the first study to show efficacy for prevention of HIV infection in a community-based cohort. During the trial, 132 participants became HIV-infected and 120 of these volunteers agreed to participate in RV152 to evaluate the effect of this primeboost regimen on clinical disease progression, immunologic and virologic outcomes. This study compared HIV-1 viral load in peripheral blood and mucosal compartments between vaccine and placebo recipients. Methods: HIV-1 viral load was measured on entry into RV152 from 42 vaccine and 58 placebo recipients paired peripheral blood (N=100) and mucosal samples [seminal plasma (SP); N=64 or cervicovaginal lavage (CVL); N=36] using the Amplicor 1.5 HIV viral load RNA assay (range: 1.70-5.88 log10 copies/ml). Results: Viral load was detected in blood (96%), SP (59%) and CVL (36%) of infected subjects. There was no difference in blood viral load between the vaccine (mean:4.05 log10 copies/ml) and placebo (mean:4.19 log10 copies/ml) groups (p=0.87). However, within the mucosal compartments, the rate of undetectable viral load was higher in the vaccine group compared to the placebo group (SP: 57% versus 28%, OR = 3.5, p=0.02; CVL: 71% versus 59%, OR = 1.7, p= 0.45; Overall: 62% versus 40%, OR = 2.5, p = 0.03). The mucosal viral load was lower in the vaccine group compared to the placebo group for SP (mean 2.22 log10 versus 2.71 log10, p=0.05) but not CVL (p=0.99) nor overall (p=0.25). Mucosal viral load was consistently lower than that of blood in both SP (mean difference=1.9 log10, p<0.0001) and CVL (mean difference=2.43 log10, p<0.0001). Conclusion: The RV144 vaccine regimen lowered the detectable rate and magnitude of viral load in SP but not blood. Ongoing studies are evaluating the robustness of this effect and potential explanatory factors such as mediating immune responses.

AIDS Vaccine 2011

161

POSTERS

Posters
P15.01

Topic 15: Prevention Strategies P15.02


Circumcision and HIV Acquisition Risk Among Men Who Have Sex with Men (MSM) in Buenos Aires, Argentina
M. Pando1, R. Marone2, I. Balan3, C. Dolezal3, V. Barreda2, A. Carballo-Dieguez3, M.M. Avila1 Centro Nacional de Referencia para el SIDA, Buenos Aires, Argentina; 2Nexo Asociacion Civil, Buenos Aires, Argentina; 3 HIV Center for Clinical and Behavioral Studies, Columbia University, New York, NY, USA
1

Men Who Have Sex with Men (MSM) from Buenos Aires, Argentina: A Suitable Group for HIV Clinical Trials
M. Pando1, M. Weissenbacher1, R. Marone2, S.M. Montano3, A. Carballo-Dieguez4, H. Salomn1, M.M. Avila1
1

Centro Nacional de Referencia para el SIDA, Buenos Aires, Argentina; 2Nexo Asociacion Civil, Buenos Aires, Argentina; 3 Naval Medical Research Unit, NAMRU-6, Lima, Peru; 4 HIV Center for Clinical and Behavioral Studies, Columbia University, New York, NY, USA
Background: For the past ten years (2000-2010), Argentinas researchers have conducted HIV-related studies among men who have sex with men (MSM) in collaboration with an MSM NGO (Nexo Asociacin Civil). This review is aimed at establishing feasibility and suitability of conducting HIV clinical trials on MSM in Buenos Aires, Argentina. Methods: Several cross-sectional studies and one follow-up cohort study were analyzed to determine HIV prevalence, incidence, and subtyping; risk factors; circumcision; behavioral factors; and willingness to participate in vaccine trials among MSM. Results: The estimated HIV prevalence has been 11-14% during the past ten years (800-1000 volunteers/year). HIV incidence, both in the cohort study and in cross sectional studies (Detune) was 4-6% persons-year. Phylogenetic analysis revealed that subtype B was the most common subtype present in MSM; yet, a higher frequency of non-B-subtypes (BF recombinants) was found in the last study conducted by the team as compared to previous studies (30.4% vs.10.5%, p<0.001). The one-year followup cohort study showed a retention rate of 91%. Studies also showed that about 50% of MSM have unprotected intercourse, 12.5% are circumcised, and 53% of men who attended the NGO for testing are repeated testers. More than 60% of MSM expressed willingness to participate in vaccine trials. Conclusion: The MSM population in Buenos Aires is very well characterized regarding HIV prevalence, incidence, molecular epidemiology, sexual risk behavior, and related parameters. Excellent retention rates were achieved in the past. Participants indicated strong commitment to participate in HIV-related studies, including vaccine clinical trials. These data clearly indicate that MSM from Argentina constitute a suitable group for HIV vaccine clinical trials.

Background: Several studies have demonstrated that circumcision reduces HIV acquisition among heterosexual men by approximately 60%.However, this may not be the case among MSM engaged in receptive anal intercourse (RAI).In Argentina, no information has been available about frequency of circumcision among MSM or its potential benefits.That was the focus of the present study. Methods: 500 MSM recruited through RDS (respondent-drivensampling) for an HIV-prevalence study were asked if they were circumcised and, if not, whether they would be interested in circumcision if it could protect them against HIV. Inclusion criteria included age at least 18 years, resident of Buenos Aires, reporting sex with men at least 10 times in their lives and at least once in the past six months. All statistics have been weighted based on the participants self-reported network size. Results: A total of 66 (14%) MSM reported being circumcised. No statistical differences were observed on age, condom-use, or sexual role (receptive or insertive with a man) between circumcised and uncircumcised. HIV prevalence was 17%. Being circumcised was not significantly associated with HIV prevalence (p=0.211). Stratifying the group according to their sexual role, 36% reported being engaged in RAI. Among those who do not practice RAI, those who were circumcised (N= 37) had no cases of HIV infection, whereas among those who were uncircumcised, 15% (40 of 265) were HIV positive (p=0.007). No difference in HIV prevalence was observed between circumcised and uncircumcised among those who practice RAI. Among those who were not circumcised,75% said that they would not be willing to be circumcised even if it could be beneficial for HIV infection risk reduction. Conclusion: Further exploration is required on the potentially protective role that circumcision may have for MSM who refrain from RAI.Attention should be paid to the lack of motivation of MSM to be circumcised for HIV-prevention.

POSTERS 162 AIDS Vaccine 2011

Topic 15: Prevention Strategies P15.03


Community Awareness About AIDS Vaccine in Georgia
N. Gulua1, O. Gerzmava2, A. Lochoshvili1, I. Norakidze1, N. Zakareishvili3 Georgia Health and Social Projects Implementation Centre, Tbilisi, Georgia; 2Grigol Robakidze University, Tbilisi, Georgia; 3 UNFPA, Tbilisi, Georgia
1

Posters
P15.04
Griffithsin, Cyanovirin-N and Scytovirin Inhibit HIV-1 Binding and Transfer via the DC-SIGN Receptor
K.B. Alexandre1, E.S. Gray1, H. Mufhandu2, P.L. Moore1, J. McMahon3, E. Chakauya2, B. OKeefe3, R. Chikwamba2, L. Morris1
1

Background: Community education for HIV/AIDS vaccine development and research and community preparedness for vaccine trials are fundamentally linked. Vaccines have already been proven to be effective and efficient means of preventing a number of other serious viral diseases. Knowledge and empowerment, through education, are the tools by which people and communities gain mastery over issues of concern. In Georgia PLWHA has lack of information on AIDS vaccine. Methods: Aim of the study was to assess community knowledge on HIV vaccine development. A study was carried out in four main cities of Georgia: Tbilisi, Kutaisi, Batumi and Zugdidi. The data were collected through administration of an anonymous questionnaire. In total 315 interviews were carried out. The interview addressed understanding of AIDS vaccine, opinions, views, fillings and target community approach. Results: Results of this survey demonstrate informational gaps in communitys knowledge of HIV vaccine and emphasize the need for training on HIV Vaccine and its development. Only 37 % had heard about an AIDS vaccine, while 63% did not have any information. Only 18% of respondents had a good understanding regarding AIDS vaccine. The main primary source regarding AIDS vaccines were 28% media and public 43%. The respondents had numerous questions regarding HIV vaccine (questions about testing, distribution, etc.) Some participants believed an HIV vaccine would be available in the future. Others believed it would not. Conclusion: The present findings suggest the importance of carefully planning and developing strategies designed to reach communities at elevated risk for acquiring HIV/AIDS in order to increase HIV vaccine knowledge, acceptability and trust and to dispel; misinformation and undue fears in regard to future HIV vaccine.

National Institute for Communicable Diseases, Johannesburg, South Africa; 2Centre for Scientific and Industrial Research, Pretoria, South Africa; 3Center for Cancer Research, Frederick, MD, USA

Background: The lectins Griffithsin (GRFT), Cyanovirin-N (CV-N) and Scytovirin (SVN) are potential microbicides that inhibit HIV1 infection by binding to mannose-rich glycans on the envelope. Since the DC-SIGN receptor plays a key role in the sexual transmission of HIV-1, we investigated whether these lectins could inhibit HIV-1 transfer and infection via this receptor. Methods: Five subtype C (COT6.15, COT9.6, Du156.12, Du151.2 and CAP63.A9J) and three subtype B (QH0692.42, PVO.4 and CAAN5342.A2) viruses were incubated with each lectin then captured with the DC-SIGN receptor on Raji/DC-SIGN cells. HIV1 capture was measured by p24 ELISA. For studies of inhibition of HIV-1 transfer and infection, the virus was either incubated with the lectin before capture on Raji/DC-SIGN cells and transfer to JC53bl-13 cells or first captured with Raji/DC-SIGN cells prior to the addition of the lectin and then JC53bl-13 cells. The inhibition of transfer was also studied in PBMCs. Results: The 3 lectins inhibited HIV-1 capture via DC-SIGN of all 8 viruses albeit at modest levels: the average for CV-N, SVN and GRFT was 52, 38 and 26%, respectively. However, all three lectins potently inhibited the DC-SIGN mediated transfer of HIV1 to JC53bl-13 cells and PBMCs. This was particularly evident when HIV-1 was incubated with the lectins after capture by Raji/ DC-SIGN cells but prior to transfer to JC53bl-13 cells. GRFT and CV-N were at least 10 times more potent in this format. A similar trend was noted for SVN although it was less potent. Lastly, the three lectins inhibited DC-SIGN binding and transfer of subtype B and C with similar potency. Conclusion: Our study suggests that GRFT, CV-N and SVN inhibit DC-SIGN-mediated HIV-1 infection and, therefore, supports further studies of these compounds for HIV-1 prevention via mucosal routes.

AIDS Vaccine 2011

163

POSTERS

Posters
P15.05

Topic 15: Prevention Strategies P15.06


Prevention of HIV-1 Sexual Transmission Using Novel Small-Molecule CCR5 Antagonists Formulated into Temperature-Sensitive Acidic Microbicide Gel
Y. Kang1, L. Chen2, X. Ding3, W. Li3, C. Lau4, X. Lu1, Z. Tan1, L. Liu1, Z. Wu5, Z. Chen1 AIDS Institute, The University of Hong Kong, LKS Faculty of Medicine, Hong Kong, China; 2Shanghai Targetdrug CO., LTD, China; 3Shanghai Institute of Planned Parenthood Research, China; 4School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; 5Center for Public Health Research, Medical School, Nanjing University, China
1

Women Fisher Folk: Recruitment, Retention and Participation in HIV Prevention Research
A. Ssetaala1, A. Nanvubya1, J. Mpendo1, G. Asiki2, J. Miiro2, L. Nielsen1, J. Seeley2, P. Kaleebu2, A. Kamali2 Uganda Virus Research Institute-IAVI HIV Vaccine Program, Entebbe, Uganda; 2MRC/Uganda Virus Research Institute Uganda Research Unit on AIDS, Entebbe, Uganda
1

Background: In sub-Saharan Africa women constitute approximately 60% of people living with HIV/AIDS. Factors that contribute to this include lack of access to essential information and reproductive health services, economic disempowerment, sexual violence and partner risk behaviours. There is an urgent need to involve women in HIV prevention research. This study explores recruitment, retention and participation in HIV prevention research among women in fishing communities in Uganda. Methods: With funding from EDCTP, Uganda Virus Research Institute and its partners conducted an 18 months prospective cohort study in 5 fishing communities along the shores of Lake Victoria in preparation for future HIV prevention research. 2,074 volunteers (men and women) aged 13-49 years were screened using demographic, medical history and risk behavior assessment questionnaires. 1,000 high risk HIV negative volunteers were enrolled and followed up every 6 months for 18 months. Results: Women constituted 51% of those screened, and 46% of those enrolled with a median age of 28 years. Approximately 64% of women had primary education, 73% were married. Majority of Women (70%) were engaged in fish processing, 10% bar attendants and 20% were unemployed. One third of the women (33%) were engaged in transactional sex. Pregnancy rates at 18 months follow up were high (22%). 80% and 73% of the women and men respectively completed all the follow up visits. Overall 23% never reached 18 months follow up, of whom 40% were women. The overall HIV incidence was 5/ 100PY, 4.6 /100PY among women and 5.3/100PY among men. Conclusion: Approximately 30% were either unemployed or in vulnerable employment exposing them to economic disempowerment and high HIV risk exposure, in urgent need of interventions. These women were willing to participate in HIV prevention research with encouraging retention rates at 18 months.

Background: The promising finding of a nucleotide analog reverse-transcriptase inhibitor tenofovir gel in protecting women from HIV infection makes microbicide an exciting preventive intervention against HIV sexual transmission in addition to vaccine. Here we report two novel small-molecule CCR5 inhibitors, TD-0232 and TD-0680, study their antiviral activity and mechanism against HIV-1 infection, and investigate the potential of TD-0232 as a microbicide. Methods: We measured the antiviral activity of TD-0232 and TD-0680 by using both HIV pseudovirus and clinical isolatebased neutralization assays. We also determined the capability of TD-0232 and TD-0680 to inhibit Env-mediated cell-to-cell transmission. The combination effect of TD-0232 and TD-0680 with other classes of HIV-1 inhibitors was analyzed. Sitedirected mutagenesis, combination assay and molecular docking were adopted to reveal the binding mode of TD-0232 and TD0680 on CCR5. The stability profile and inhibitory activity of TD-0232 in a formulation of temperature-sensitive acidic gel were also investigated. Results: TD-0232 and TD-0680 specifically inhibited R5tropic HIV-1 and SIV strains and TD-0680 consistently had a higher potency. They also blocked cell-associated virus fusion and transmission effectively. TD-0232 showed synergy with nevirapin, whereas TD-0680 showed synergy with azidothymidine, efavirenz and tenofovir. Rhesus CCR5 was more sensitive to TD-0680, which made it suitable for in vivo monkey model studies. Homology modeling of antagonist-bound human CCR5 and antagonism in combination assay revealed that TD0232 and TD-0680 had overlapping but distinct binding modes. Furthermore, TD-0232 gel could rapidly release the drug vehicle and stably exert antiviral activity for a long term which was not affected by semen. Conclusion: TD-0232 and TD-0680 are potent inhibitors against R5-tropic HIV-1 infectivity but with distinct binding profiles on CCR5. TD-0232 is also a promising candidate for preventive microbicide.

POSTERS 164 AIDS Vaccine 2011

Topic 15: Prevention Strategies P15.07


Recruitment and Retention of HIV Serodiscordant Couples in a Pilot Daily and Intermittent PreExposure Prophylaxis Study in Masaka, Uganda
F.M. Kibengo , E. Ruzagira , A. Bwanika , U. Bahemuka , D. Katende1, A. Abaasa1, F. Priddy2, A. Kamali1
1 1 1 1 1

Posters
P15.08
Project VOGUE: HIV Vaccine Education in an Underserved Racial/Ethnic MSM Population
C.A. Bunce1, S.D. Fields1, S. Wallace2, D. Humes3, M.C. Keefer1 University of Rochester, Rochester, NY, USA; 2MOCHA, Rochester, NY, USA; 3HIV Vaccine Trials Network- Legacy, Seattle, WA, USA
1

Medical Research Council/Uganda Virus Research Institute, Kampala, Uganda; 2International AIDS Vaccine Initiative, New York, NY, USA

Background: Pre-Exposure Prophylaxis (PrEP) using antiretroviral drugs is one of the HIV prevention technologies currently being explored.High volunteer recruitment and retention rates are critical for the conduct of HIV Pre-Exposure prophylaxis trials. We describe screening, recruitment and retention of volunteers in HIV sero-discordant couple relationships participating in a pilot study of daily and intermittent PrEP in Masaka, Uganda Methods: Volunteers were recruited from an ongoing HIV serodiscordant couples HIV incidence cohort study. Enrolled volunteers were randomized to daily FTC/TDF or placebo, or intermittent FTC/TDF or placebo in a 2:1:2:1 ratio. Volunteers were reviewed weekly for the first two weeks and again 2 weeks later. Thereafter, monthly visits were conducted for 4 months and then a final post-trial drug visit 2 months after the end of the intervention period.Procedures conducted at every monthly study visit included; HIV and pregnancy tests, HIV (pre- and post-test), family planning,study medication adherence counselling, and clinical and laboratory safety assessments. Results: A total of 133 HIV serodiscordant couples were screened between September 2009 and March 2010. Of these, 72 (50% F-M+) were enrolled, 45 (34%) screened out while 16 (12%) met the eligibility criteria but were not enrolled because study accrual had been achieved. The main reasons for screen failure were: low creatinine clearance (57.8%), abnormal urinalysis results (15.6%), breastfeeding (4.4%) and receipt of ART by HIV-infected partners (4.4%). Each of the remaining 8 volunteers (17.8%) had a different individual reason for screen failure. Sixty eight (94%) volunteers completed the study while 4 dropped out due to pregnancy (3) and previously undiagnosed Hepatitis (1) Conclusion: We successfully recruited and retained heterosexual HIV discordant couples in this pilot daily and intermittent PrEP study. Laboratory abnormalities were responsible for most screen failures while drop-outs were mostly due to pregnancy

Background: African-American and Latino men who have sex with men (MSM) are currently the most at risk for becoming infected with HIV among all racial sub-populations of MSM. The House/Ball community is a subset of this population and at particularly high risk related to unprotected anal sex and substance abuse within their closed social network. Rochester/ Buffalo area has approximately 15 houses, with 10-12 members in each, age range 15-45. The House/Ball community has not been targeted for HIV prevention interventions/programs including interventions regarding participation in HIV vaccine clinical trials and other bio-medical research. Methods: Project VOGUE, a 2-year initiative funded by the Legacy Project, consisted of community based participatory research (CBPR) principles; as well as education about HIV prevention and vaccine trials to the House/Ball community. Qualitative interviews and focus groups were conducted. A train the trainer session was held with 13 key informants that resulted in a final curriculum. A five session group level intervention resulted, consisting of substance abuse, HIV prevention/transmission, partner violence, HIV vaccine clinical trials, and community resources. Knowledge; skill building exercises; a community change project by the attendees; and final session to evaluate the process through a post- questionnaire/knowledge assessment was conducted. Results: The leaders of each house joined to form the Council of Houses (COH) as expert informants. The COH members have been engaged in the process and recognize the need for HIV prevention/vaccine research. The COH also developed governance structures that frame, inform, and influence the behavioral social norms of the local House Ball community. Conclusion: Expert informants and CBPR methods proved effective in developing a project to educate MSM involved in the House Ball community about HIV vaccine research. This model can be piloted for reaching other hard to reach populations. Future research to measure the sustainability and effectiveness of such interventions need to be conducted

AIDS Vaccine 2011

165

POSTERS

Posters
P15.09

Topic 15: Prevention Strategies P15.10


Evaluating Predictors of HIV Infection Among MSM in Nigeria Towards Participation in HIV Prevention Vaccine Trial
A. Adeyemi1, K. Oyediran1, O. Fakunle2, K. Issa3, A. Azeez3
1 MEASURE Evaluation, Asokoro Abuja, Nigeria; 2University College Hospital, Ibadan, Nigeria; 3Federal Ministry of Health, Abuja, Nigeria

The Renewed Canadian HIV Vaccine Initiative


N. Varughese1, P. Milsom1, L. Jonsdottir1, S. Sternthal1
1

Government of Canada, Ottawa, Canada

Background: To renew the Canadian HIV Vaccine Initiative (CHVI) and to align CHVI activities with the priorities of the Global HIV Vaccine Enterprises 2010 Scientific Strategic Plan (SSP). Methods: The renewal of the CHVI incorporated: the findings of international policy consultations; analysis of the changing HIV vaccine research and development landscape; consultations and emerging priorities of the 2010 Global Enterprises SSP; and findings from Canadian stakeholders to inform the implementation of the CHVI Research and Development Alliance. Results: In July 2010, the Government of Canada and the Bill & Melinda Gates Foundation renewed the CHVI, with the objective to further strengthen global efforts to accelerate the development of HIV vaccines and to contribute to the achievement of the Global HIV Vaccine Enterprises Scientific Strategic Plan (SSP). Key areas of focus are: Advancing Basic Science; Translating Basic Science into Clinical Trials; Addressing Enabling Conditions; Preventing Mother-to-Child Transmission of HIV; and Supporting Coordinated Efforts. The cornerstone of the renewed Initiative is the establishment of a CHVI Research and Development Alliance, based in Canada to serve as the focal point for Canadian research, product development and technical expertise related to HIV vaccines. The Alliance will be linked to Low-to-Middle Income Countries research institutions particularly in Africa to leverage local expertise and knowledge, build on cooperative research and policy activities, and expand needed local capacity. The Alliance will be supported by a coordinating office and guided by a new Advisory Board. Conclusion: In moving forward, the Alliance will build on Canadian strengths in HIV vaccine research and development. The renewed CHVI will enable Canada to be a leading contributor to global efforts in developing a safe, effective, affordable and globally accessible HIV vaccine, and will help cement Canadas place in the world as a leader in HIV vaccine research.

Background: Men who have sex with men (MSM) have a high risk of HIV acquisition worldwide. Effective prevention strategies are needed among this group. They are hard-to-reach population and are often neglected in evidence-based prevention programs. Importantly, they can benefit from HIV vaccines and could participate in HIV prevention trials. The paper evaluates the predictors of HIV infection among MSM in Nigeria. Methods: Secondary data analysis of a cross-sectional survey among MSM in six Nigerian states was done. Questionnaires were administered to 1545 MSM from a respondent-driven sampling with age range 18 49years. Multivariate logistic regression models were used to evaluate risk factors to HIV infection among the MSM. Results: HIV prevalence among MSM was 17.2% with state HIV prevalence being 5.7% for Cross rivers state; 9.6% Oyo; 11.4% Kano; 23.1% Kaduna; 27.1% Lagos and 44.4% in FCT. The mean age was 25.46.0years; 71.9% were currently married; median anal sex partner in the last 6months was 3; those that were tested and received their HIV result in the last 12months were 31.5%; condom use at last anal sex: 52.1% and 41.9% had sex with girlfriends. The significant predictors of HIV were being a paid partner OR 1.4 (95% CI 1.1 -2.3), anal sex without condom OR 1.9 (95% CI 1.3 -2.9; having 3 partners OR 1.5 (95% CI 1.1 -2.6) while tertiary education was protective with OR 0.6 (95% CI 0.4 -0.9). Conclusion: Low condom uptake and paying for anal sex have serious implications in HIV prevention among MSM. Similarly, coordinated and effective prevention interventions are highly needed among them because their HIV prevalence is much higher than that of the national prevalence of 3.6% in Nigeria. These prevention efforts include partner reduction, uptake of HIV testing and promotion of condom use in any form of sexual relationships.

POSTERS 166 AIDS Vaccine 2011

Topic 15: Prevention Strategies P15.11


Attitudes and Intent to Use PrEP Among Current Phase II Preventive HIV-1 Vaccine Trial Participants
T. Madenwald1, J. Fuchs2, M. Sobieszczyk3, S. Karuna1, A. Sherwat4, B. Koblin5, G. Broder1, N. Eaton1, M. Andrasik1, S. Hammer3, Division of AIDS (DAIDS)HIV Vaccine Trials Network6
1

Posters
P15.12 LB
Ring Formulated NNRTI 5-Chloro-3(Phenylsulfonyl)Indole-2-Carboxamide (CSIC) Showed No Toxicity in Rhesus Monkeys and Retained Anti-Viral Activity
P. Gupta1, P. Marx2, J. Moss3, A. Smith3, D. Ratner1, M. Hunter2, P. Tarwater4, L. Rohan1, A.M. Cole5, M.A. Parniak1 University of Pittsburgh, Pittsburgh, PA, USA; 2Tualne National Primate Reserach Center, Covington, LA, USA; 3 Auritec Pharmaceuticals, Santa Monica, CA, USA; 4Texas Tech University Health Sciences, El Paso, TX, USA; 5University of Central Florida, Orlando, FL, USA
1

HIV Vaccine Trials Network, Fred Hutchinson Cancer Research, Seattle, WA, USA; 2San Francisco Department of Public Health, San Francisco, CA, USA; 3Columbia University, New York, NY, USA; 4National Institute of Allergy and Infectious Diseases, Division of AID, Washington, DC, USA; 5New York Blood Center, New York, NY, USA; 6National Institute of Allergy and Infectious Diseases, Washington, DC, USA

Background: In November 2010, the iPrEx study reported that pre-exposure prophylaxis (PrEP) with daily tenofovir/ emtricitabine reduced HIV infections by 44% among men who have sex with men (MSM) and transgender (TG) women. To inform future HIV vaccine trial design, MSM and TG phase II vaccine trial participants were asked about perceived significance of these results, intent to use and access to PrEP, and impact on trial recruitment and retention. Methods: HVTN 505 is a Phase II, randomized, placebocontrolled HIV vaccine trial enrolling 1350 healthy, at risk HIVuninfected MSM and TG women at 21 sites in the United States. From January-March 2011, HVTN 505 participants completed an optional, anonymous web-survey during their regularly scheduled study visits. Results: Of the 487 participants who had a visit during the survey time period, 354 completed the survey; 82% were white, and 73% had health insurance. Overall, 31% thought it moderately or very likely they would take PrEP in the next year, while 39% stated they werent likely to take it at all; 54% and 53% would be very likely to take PrEP if made available through a clinical trial or covered by their health insurance, respectively; 68% were not likely at all or slightly likely to take PrEP at their own expense. Most (90%) believed taking PrEP would not change their willingness to stay in HVTN 505, while 44% thought it may affect whether others would enroll. Overall, responses to open-ended questions revealed positive perceptions of iPrEX results, concerns about access and affordability, and continued commitment to vaccine trial participation. Conclusion: Intent to use PrEP was modest among survey respondents enrolled in HVTN 505. While concerns about access may limit PrEP use, commitment to continued participation in 505 in the setting of PrEP is high. Enrolled trial participants can provide important formative input about emerging prevention technologies.

Background: CSIC is an NNRTI with potent antiviral activity in cell culture against cell-free and cell associated HIV-1.. and demonstrated a potent memory or protective effect against HIV infection. In this study CSIC has been evaluated for its cytotoxicity and antiviral activity in a cervical tissue derived organ culture. CSIC formulated as a vaginal ring was assessed for in vivo toxicity in rhesus macaques (RhM) as well as antiviral activity in an ex vivo challenge experiment against RT-SHIV. Methods: CSIC was evaluated in organ culture for toxicity and its ability to block HIV-1 transmission across cervical mucosa. Silicone rings containing CSIC were applied intravaginally in 4 RhM for 14 days. Every three days, blood and cervicovaginal lavage (CVL) samples were analyzed for serum CSIC levels and for CVL antiviral activity. Vaginal and cervical biopsies taken prior to and fourteen days post-application of the ring were subjected to challenge with RT-SHIV in an ex vivo organ culture model. Results: Unformulated CSIC blocked transmission of both R5 and X4 HIV-1 across cervical mucosa in our ex vivo organ culture model. Furthermore, CSIC did not induce proinflammatory cytokine response in tissues up to 72 hr after exposure. All CVL samples collected from the animals during CSIC ring exposure contained substantial amounts of CSIC and were found to have high antiviral activity. There was no toxicity observed from CSIC formulated rings during vaginal exposure. Importantly, cervical and vaginal biopsies from CSIC-instilled animals prevented RTSHIV transmission in ex vivo organ culture compared to preexposure base line samples. Conclusion: CSIC showed potent antiviral activity with no detectable cytotoxcity in vitro and ex vivo organ culture. The presence of antiviral CSIC in CVL and cervical/vaginal tissues with no toxicity throughout a fourteen day exposure period in vivo suggests that ring-formulated CSIC warrants further development as a topical microbicide.

AIDS Vaccine 2011

167

POSTERS

Posters
P15.13 LB

Topic 15: Prevention Strategies

Knowledge of HIV/AIDS Transmission, Prevention, Treatment and Sexual Behaviour among PLWHA in Eastern Uganda
J. Abisa 1
1 Makerere University College of Health Sciences, Kampala, Uganda

Background: Individual sexual behavior and knowledge about HIV are major determinants of HIV prevalence in a population. HIV among adults is mainly transmitted through heterosexual contacts. Most prevention programs in Uganda therefore focus on strategies to reduce sexual transmission through the ABC strategy.This study presents findings from data collected from 4 districts of Eastern Uganda in 2009 to explore knowledge of HIV/ AIDS transmission, prevention, treatment and sexual behaviour among PLHIV. Methods: An LQAS baseline survey was conducted during November and December 2009 in 4 districts namely; Busia, Butaleja, Pallisa and Sironko. Nineteen PLHIV were drawn from PHA Organizations registers from each of the 4 districts giving a total sample of 76. The LQAS baseline survey was conducted for selected HIV/AIDS, TB and Malaria indicators. Results: Seventy Six percent of the PLHIV were women, 28% were male. Most of PLHIV interviewed were aged 35-44 (48%) and a majority were Widowed (43%). About 41% had not completed primary level education. On levels of knowledge of prevention, abstinence was mentioned by at least 90% of PLHIV, condom use was reported by 95%. While 71% rejected that HIV can be transmitted through a mosquito bite, 90% rejected that the virus can be transmitted through sharing utensils. However although 67% rejected Witchcraft, 28% accepted it as a transmission mode. At least 90% mentioned MTCT as another mode while 92% knew where to access information Conclusion: The survey findings show that although knowledge of HIV/AIDS transmission, prevention and treatment among PLHIV is very high, PLHIV still engage in risky sexual behavior. This is likely to fuel drug resistance to the first line treatment currently being used in the country, thereby defeating the current secondary prevention programs. There is need to initiate programs that will help to transform high levels of knowledge into behavior change among PLHIV.

POSTERS 168 AIDS Vaccine 2011

Topic 16: Social/Ethical/Access/Regulatory Issues P16.01


HIV Vaccine Acceptability and Behavioural Risk Compensation Among Men Who Have Sex with Men in a Resource-Poor Setting in Africa
O.A. Busari1, N. Martha2, A. Busari3, S.M. Agboola1
1

Posters

P16.02
HIV and STI Prevalence Among Men Who Have Sex with Men Recruited Through Respondent Driven Sample in a Nigerian Oil-Rich Port Harcourt City, Nigeria
O.A. Busaro1, M. Nakayima2, A. Adeyemi3, S. Agboola1
1

Federal Medical Centre, Ido-Ekiti, Nigeria; 2The AIDS Support Organization, Masaka, Uganda; 3CARE AIDS International, Ado-Ekiti, Nigeria

Federal Medical Centre, Ido-Ekiti, Nigeria; 2The AIDS Support Organization, Masaka, Uganda; 3Family Health International, Abuja, Nigeria

Background: The Thailand phase-3 HIV vaccine trial two years ago raises hope of a future effective vaccine. But the effective roll-out and use of a future HIV vaccine depends largely on its acceptability. We assessed acceptability of HIV vaccines and behavioural risk compensation among men who have sex with men (MSMs) in a cosmopolitan high density sub Sahara African city, Lagos, Nigeria Methods: A structured questionnaire was designed based on formative research among some MSMs, HIV physicians and HIV preventive researchers in Nigeria. It was programmed on laptop computers and administered by trained interviewers. Participants were recruited using venue-based sampling from sex venues, public motor parks and garages, and inner-city club houses in Lagos. We assessed HIV vaccine acceptability using conjoint analysis and a factorial experimental design, and risk behaviour intentions in response to HIV vaccine use Results: Participants were 103 MSM; mean age = 30.4 (SD = 4.7) years. Only 9 (8.7%) engaged in paid sex. Had sex with average of 3.4 male partners in the last 6 months. More than half reported STI diagnosis in the last 1 year.HIV vaccine acceptability ranged from 87.9 (SD =25.2) and 42.3 (SD = 36.5) on a 80-point scale. Vaccine induced seropositivity has the most impact on acceptability (27.4; p<0.001), followed by efficacy (22.1; p <0.001), side effects (11.5; p = 0.002), duration (8.3; p = 0.002, and out-of-pocket cost (6.2; p = 0.002). 60 (58.3%) reported intention to increase sexual risk behaviour after HIV vaccine Conclusion: Study confirms and support need for development of interventions to reduce impact of vaccine induced seropositivity, enhance acceptability of future vaccine, and improve understanding of possible side effects which may facilitate HIV vaccine uptake among MSM and other vulnerable groups in Nigeria and West African subregion. Finally behavioural interventions to reduce risk conpensation remains a critical aspect of

Background: MSMs are one of the high risk populations for HIV and STI and a potential cohort for HIV vaccine trials There is no prevalence study of MSMs in West Africa. Respondent driven sampling (RDS), a novel methodology designed to access hidden populations was used for the first time in West Africa to recruit MSM Methods: The recruitment for the study started with the selection of 10 first generation participants (seeds) with potential to track fellow MSMs through their networks. Criteria for recruitment included: residency of Port Harcourt, Nigeria; age 18 years; self report of having sex with men at least 8 times in their lives and at least once in the last six months. Participants were screened for HIV and STI. Data were analyzed with RDS Analysis Tool (RDSAT) Results: 98 MSMs were recruited through RDS for the study. Prevalence of HIV, HBV and HCV in the participants was 12.1%, 27.7% and 10.3 %respectively. 57 (55.9%) of the participants had HPV infection. Conclusion: This is perhaps the first research study of MSMs in West Africa. Results show a high prevalence of HIV, HBV, HCV and HPV co-infections in MSM population. This lends support to MSMs as potential participants for HIV vaccine trials and also a target population for future use of the vaccine when it eventually becomes available.The study also revealed that RDS methodology helps recruit a diversity of MSMs, usually missed with other recruitment methods

AIDS Vaccine 2011

169

POSTERS

Posters
P16.03

Topic 16: Social/Ethical/Access/Regulatory Issues P16.04


Preparing for the Availability of a Partially Effective HIV Vaccine
R.T. Chen1, C. Hankins2, S. Osmanov3, J. Esparza4, N.L. Michael5, M. McCluskey6, M. Warren7 Centers for Disease Control and Prevention, Atlanta, GA, USA; 2UNAIDS, Geneva, Switzerland; 3WHO, Geneva, Switzerland; 4Bill & Melinda Gates Foundation, Seattle, WA, USA; 5U.S. Military HIV Research Program, Rockville, MD, USA; 6U.S. Military HIV Research Program, Washington, DC, USA; 7AVAC, New York, NY, USA
1

Are Adults in Soweto Still Willing to Participate in Future Vaccine Trials? A Qualitative Study
J. Dietrich1, P. Maesela1, A. Kagee2, G. Gray1
1 2

Perinatal HIV Research Unit, Johannesburg, South Africa; Stellenbosch University, Cape Town, South Africa

Background: South Africa bears the greatest burden of HIV/ AIDS globally with a prevalence rate of 10.9%. The search for an effective preventative HIV vaccine is therefore ongoing. A potential candidate vaccine will be tested amongst high risk populations in phase II and III HIV vaccine trials. Large numbers of HIV-negative volunteers will be required to enroll and return regularly for assessment over a number of years. This study explored willingness of adults to participate in future preventative vaccine clinical trials. Methods: Thirty HIV negative participants, aged 18-35 years, were recruited via the Voluntary Counselling and Testing site at the Perinatal HIV Research Unit in Soweto, Johannesburg. A qualitative methodology was used to collect data via in-depth interviews. Participants were allowed to use their language of choice. Interviews were recorded using digital recorders. Interviews were then transcribed verbatim and translated to English. Data analysis was conducted using Maxqda, a qualitative software data management and analysis programme. Results: The main themes were: knowledge about vaccines, enablers and inhibitors to participation in vaccine trials. Vaccine knowledge was variable. Those with good knowledge had heard about trial participation from friends who had participated in clinical trials. Enablers included having enough time to make the decision to participate, treatment for side effects and partner and parent approval as well as support. Inhibitors were: being stigmatized as HIV positive, experiencing side effects, having misconceptions about vaccine trials, misconceptions about the intentions of the research unit, parent and partner disapproval, knowing friends who were HIV positive and knowing friends and family who had died of HIV/AIDS. Conclusion: Providing the correct information about vaccine trials and involving significant others are important when inviting volunteers to participate in future vaccine trials. This may translate to involving significant others in the information provision sessions prior to participation.

Background: A satellite symposium held at AIDS Vaccine 2010 updated thinking on downstream implementation issues for a partially effective HIV vaccine. The session provided the first comprehensive discussion since 2002 when CDC and WHO anticipated the results of the first Phase 3 HIV vaccine (VaxGen gp120) trial. Methods: Presentations in 2010 included: 1) Results of mathematical models (some with health economic parameters) from different research groups applying a priori consensus RV144 trial results to Thailand, South Africa, U.S., and Australia and 2) Relevant lessons from implementation of hepatitis B vaccine (HBV), human papillomavirus (HPV) vaccine, antiretroviral preexposure prophylaxis (PrEP), annual influenza vaccine strain selection, and post-RV144 planning in Thailand. A moderated discussion with audience members focused on developing and developed world considerations. Results: Issues to emerge included: 1) All mathematical models projected a substantial number of HIV infections averted with the RV144 vaccine, especially if boosters turn out to be efficacious. 2) HBV vaccine introduction in almost all countries initially targeted high-risk populations given higher cost-effectiveness, however, acceptably higher uptake was attained only after change to universal infant vaccination policy combined with affordable vaccines. 3) Attempts to combine universal HPV vaccine introduction in adolescents in the US with school entry requirements was unsuccessful due to concerns about inadequate safety data. 4) Should future HIV epidemiology in the U.S. warrant recommendations for routine childhood HIV vaccination, the Vaccine For Children program provides a stable funding mechanism for those otherwise without health insurance. 5) Many of the challenges with annual influenza vaccine strain selection may be magnified with efforts to arrive at annual consensus HIV strains. 6) Globally, infant immunization programs are well-established while those for adolescents or adults are not. 7) Prioritization vs. competing priorities and access/affordablity are also concerns. Conclusion: This satellite provides a useful start on planning for complex HIV vaccine implementation issues.

POSTERS 170 AIDS Vaccine 2011

Topic 16: Social/Ethical/Access/Regulatory Issues P16.05


HIV Vaccine Knowledge, Concerns and Misconceptions Among Vulnerable Communities in Resource-Poor Settings of Two Adjoining Border Cities in Africa
O.A. Busari1, S. Agboola1, A. Busari2, M. Nakayima3, A. Adeyemi4 Federal Medical Centre, Ido-Ekiti, Nigeria; 2CARE AIDS International, Ado-Ekiti, Nigeria; 3The AIDS Support Organization, Masaka, Uganda; 4Family Health International, Abuja, Nigeria
1

Posters

P16.06
Sustaining Community Interest on the Road Towards an HIV Vaccine
S. Sigirenda1, W. Kidega1, L. Nielsen 1
1

International AIDS Vaccine Initiative, Kampala, Uganda

Background: With the increased hope of availability of future effective HIV vaccine after the modest success of the Thai phase-3 HIV vaccine trial last year, it is important to start early preparation for effective distribution and uptake particularly among vulnerable communities in sub-Saharan Africa at highest risk for HIV. The study was to identify and assess knowledge, beliefs and concerns about future HIV vaccines among adults in vulnerable communities in resource-poor settings of Lagos and Cotonou in West Africa Methods: A cross-sectional survey (based on qualitative findings) was conducted among 615 adults ( 18 years) in two border communities of Lagos, Nigeria, and Cotonou, Benin, both in the West African subregion. They were recruited using multisite venue based sampling. Median age was 26 years; 42% were females; and average annual income was <300USD per annum. The survey characteristics included HIV vaccine knowledge, beliefs and concerns, and the state/quality of (or access to) health services in their communities Results: Study revealed majority of participants (87%) never heard of HIV vaccines and almost three-quarters (74%) were concerned they might contract HIV from a vaccine. Concerns about HIV vaccine-related stigma and discriminations were shown by about 68%. Half reported concerns about confidentiality and most of the participants (97%) confirmed no access to quality health services in their community Conclusion: Lack of knowledge and concerns, misconceptions and mistrust among vulnerable communities in resource-poor settings present critical challenges to HIV vaccine distribution, and may hinder the effectiveness of vaccine in controlling AIDS epidemic. Culturally appropriate, empirically based individualand community-level educational and social interventions and outreach may be vital to address HIV concerns and misconceptions so as to achieve successful dissemination

Background: After years of conducting HIV Vaccine research, the hope of finding an HIV Vaccine has been rekindled by recent promising results on proof of concept and broad neutralizing antibodies. Community involvement is important for continued support of the research process. This abstract presents views of community members surrounding a research site in Entebbe Uganda on sustaining community interest for HIV Vaccine research collected in a qualitative study. Methods: Focus group discussions and Key informant interviews were held with fourteen community members. Questions were based on a broad question; In the absence of an effective vaccine how can community interest be sustained? Participants were chosen purposively based on their involvement in community engagement and were gender balanced. Content and thematic analyses were conducted to get the final results. Results: Community members appreciated scientific efforts to develop a vaccine. Most participants believed that a genuine partnership between researchers and communities was key to progress towards an HIV Vaccine. Key strategies were related to; - Instill a sense of hope: Research teams should draw from the success of the polio and measles vaccines, and explain recent advances in order to demonstrate that there is renewed hope that an HIV vaccine is possible - Simplify communications: At all levels of conducting research information dissemination, priority should be given to simplifying scientific terms to contribute to sustaining community support. - Community education: Innovative stakeholder education ensures ongoing support, increasing levels of study participation. Conclusion: More than ever before, sustaining community interest in HIV Vaccine research requires instilling hope,communication and stakeholder support. This involves a number of innovative, adaptive, context driven strategies to ensure community support goes hand in hand with the research process. The recent promising results show that a vaccine is possible.

AIDS Vaccine 2011

171

POSTERS

Posters
P16.07

Topic 16: Social/Ethical/Access/Regulatory Issues P16.08


Sustaining Scientific Momentum in an Uncertain Global Economic Environment: The HIV Vaccine Funding Landscape in 2010
K.A. Fisher1, C. Avila2, B. Gobet2, T. Harmon3, P. Harrison1, R. Lande4, W. Lee1, W. Muchenje1, M. Warren1 AIDS Vaccine Advocacy Coalition, New York, NY, USA; UNAIDS, Geneva, Switzerland; 3International AIDS Vaccine Initiative, New York, NY, USA; 4International Partnership for Microbicides, Silver Spring, MD, USA
1 2

Community Engagement in HIV Vaccine Research: A Multiple Embedded Case Study in Canada, India, South Africa and Thailand
P.A. Newman1, G. Lindegger2, V. Chakrapani3, S. Roungprakhon4, C. Slack2, Z. Essack2, J. Koen2, M. Shanmugam3, S. Tepjan1, C. Logie1
1

University of Toronto, Toronto,ON, Canada; HAVEG, School of Psychology, University of KwaZulu-Natal, Pietermaritzburg, South Africa; 3Indian Network For People Living With HIV/AIDS (INP+), Chennai, India; 4Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
2

Background: The need for community engagement in HIV vaccine research has become a mantra among populations at higher risk of HIV exposure, people living with HIV, advocacy groups, community-based, non-governmental and government organizations, clinical trialists and researchers across low-andmiddle-income (LMIC) and high-income countries. Nevertheless, scant research has identified and assessed stakeholder roles and challenges for community engagement in diverse country settings. Methods: We implemented an embedded exploratory case study with a multiple case design. We conducted in-depth, semistructured interviews with persons from populations at higher risk of and living with HIV, community advocates, service providers and HIV experts in Ontario, Canada; Chennai, India; Johannesburg, South Africa; and Bangkok and Chiang Mai, Thailand, and community focus groups. The central questions were: What are appropriate roles of community stakeholders in HIV vaccine trials? What are the challenges and benefits of community engagement? Data were analyzed across and within multiple stakeholder groups and countries using narrative techniques and a constant comparative method. Results: We conducted 92 interviews and 10 focus groups (n=91) across 4 countries (N=183); 55% men, 43% women, 2% transgender women. Cross-cutting themes included altruism; challenges in identification of genuine community representatives; trial literacy; mistrust of medical research; late invitation to engage in already-planned trials; lack of specificity on appropriate roles for community stakeholders; dealing with negative trial results; and dissemination of information. In LMIC sites, economic disparities between trial sites and sponsor nations, and sustaining support for community-based organizations beyond trial duration emerged as important challenges. Distinct local vaccination cultures arose in each country. Conclusion: Meaningful community engagement challenges us to: identify appropriate roles for community stakeholders commensurate with time and expertise; engage early in the trial planning process; maintain transparency in information shared and bridge siloization of knowledge; and to support capacitybuilding and sustainable community infrastructure despite the episodic implementation of clinical trials.

Background: Since 2004, the HIV Vaccines and Microbicides Resource Tracking Working Group has employed a comprehensive methodology to track resource trends in R&D for new HIV prevention options -- vaccines, microbicides, pre-exposure prophylaxis (PrEP), adult male circumcision and others. Information on investment is critical to identify trends in investment and provide a fact base for policy advocacy. Methods: To estimate annual investment in HIV vaccine R&D, data were collected from government agencies, nonprofit research organizations, foundations, and pharmaceutical/biotechnology companies on annual disbursements for product development, clinical trials and trial preparation, community education, and policy advocacy efforts. Results: Preliminary estimates suggest funding for HIV vaccine research in 2010 continues a decline from its peak in 2007, with a slight decrease in funding by the U.S. government, by far the largest supporter of HIV vaccine research. European public sector funding for HIV vaccine research also continued its negative trend of the last two years. The Bill & Melinda Gates Foundation continued to provide the great majority of philanthropic funding for the HIV vaccine field, but at lower levels in 2010 due to funding cycles. 2010 also saw a renewed effort by a number of commercial entities in HIV vaccine research, primarily in the biotechnology industry, but their funding contributions remain quite small in comparison to public and philanthropic sector support. Conclusion: Scientific momentum in the HIV vaccine R&D field continued in 2010, with the discovery of additional neutralizing antibodies, new work on mosaic gene vaccine design, crucial analysis of positive results stemming from the RV144 vaccine trial, and preparations for follow-up trials over the next five years. A continued downward or flat-funding trend at a time of public sector austerity could present difficult choices for the field, especially given the resource requirements for expensive clinical follow-up

POSTERS 172 AIDS Vaccine 2011

Topic 16: Social/Ethical/Access/Regulatory Issues P16.09


A Social Vaccine? Social and Structural Correlates of HIV Vaccine Acceptability among Key Populations at Higher Risk of HIV Exposure in Thailand
P.A. Newman1, S. Roungprakhon2, S. Tepjan3, S. Yim3
1

Posters

P16.10
Community Conversations Around Good Participatory Practice Guidelines: The Need for Dialogue Between Communities, Governments and Researchers
G. Calazans1, J. Beloqui2
1 So Paulo HVTU - CRT-DST/Aids, So Paulo, Brazil; 2GIV Grupo de Incentivo Vida, So Paulo, Brazil

University of Toronto, Toronto, ON, Canada; 2Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand; 3 Factor-Inwentash Faculty of Social Work, Toronto, Canada
Background: The effectiveness of vaccines in reducing new HIV infections is contingent on their acceptability to end-users; yet limited investigations have addressed social- and structural-level factors that may influence acceptability. We explored social and structural factors associated with HIV vaccine acceptability in Thailand, a nation well-positioned to be among early adopters of a future vaccine. Methods: From 2006-2009 we conducted a mixed methods investigation: Phase 1 (n=39)in-depth, semi-structured 1-hour interviews in four Thai cities among key populations at higher risk of HIV, community service providers and HIV experts recruited using purposive sampling. Interviews were digitally recorded, transcribed, translated into English, and analyzed using narrative techniques from grounded theory; Phase 2 (n=326)intervieweradministered survey programmed on laptop computers among participants recruited using venue-based sampling. Results: Among 365 participants, 66% were men, 22% women, 12% transgender women; mean age = 28 years. Cross-cutting social and structural correlates of HIV vaccine acceptability included: social saturation; peer, familial and societal stigma; discrimination in healthcare; and vaccine cost. Populationspecific challenges included: 1) gay men/MSM: anti-gay prejudice, being outed, and behavioral disinhibition; 2) transgender women: transphobia, being grouped with MSM, and challenges in condom negotiation with straight men; 3) male and female sex workers: primary partners mistrust; new challenges to enforcing condom use with male clients; and criminalization of sex work; and 4) injection drug users: lack of social and community support; criminalization of drug use, and lack of harm reduction approaches. Conclusion: Population-specific and cross-cutting social and structural factors may place constraints on HIV vaccine uptake among key populations at higher risk for HIV in Thailand. Complementing HIV vaccine roll-out with social- and structurallevel interventions that produce an enabling environment encouraging social support for vaccination; combatting HIV and anti-gay stigma; subsidizing vaccine costs; and decriminalizing adult sex work and drug usemay optimize the effectiveness of vaccines in controlling HIV in Thailand.

Background: A partnership between the So Paulo HVTU and the Brazilian NGO GIV has gathered Brazilian community members to discuss the Good Participatory Practice (GPP) Guidelines in a project funded by AVAC. The objective was to insist on the need to develop a national document to establish guidelines and regulations on participatory practices and create a favorable context to support the document and its implementation. Methods: Brazilian community members strategic for HIV vaccine trials were invited to a one-day consultation meeting. Some have already participated in a previous consultation meeting held in 2008 and others were new to GPP, though engaged in LGBT and HIV/Aids activism and/or AIDS programs at local, state and national levels. Presentation was developed on the results from the 2008 GPP consultation and its referrals, GPP Revision process and local consultations held in the South and Northeast of the country. Questions were raised to promote the debate around the steps needed for the implementation of a national document on GPP. Results: Brazilian stakeholders recognized that national GPP guidelines for health research should be implemented. To allow implementation of such guidelines it was pointed that there should be: a link with the national ethical framework, though recognizing the need for more thorough mechanisms of monitor of daily trial activities by communities and register of volunteers experience, and problems they face; community education efforts developed in order to promote a minimum understanding on research processes and organize a campaign in health services to instruct people on what institutions should be seen if facing problems in trials; strategies to engender opportunities for dialogue between scientific community and civil society so that control does not depend only on external bodies. Conclusion: To ensure implementation of Brazilian national GPP guidelines there is a need to strengthen dialogue strategies between community representatives, governmental stakeholders and researchers around participatory practices.

AIDS Vaccine 2011

173

POSTERS

Posters
P17.01

Topic 17: T Cell Immunity P17.02


Frequency of CD4+ and CD8+ IL17-Secreting T Cells During Acute/Early HIV Infection and in Chronic Aviremic HIV Controllers
G. Turk1, Y. Ghiglione1, M.E. Socias2, A.M. Rodriguez1, J. Favilene1, M.J. Ruiz1, O. Sued3, D. Pryluka4, N. Laufer1, P. Cahn2, M.M. Gherardi1, H. Salomon1
1 2

Synergistic Upregulation of Systemic IL-10 and Functional Enhancement of CD4+ T Cells by IL-10 Receptor Blockade in HIV+/MTB Co-Infection
S. Chetty1, F. Porichis2, P. Govender1, J. Zupkosky3, M. Pillay1, B.D. Walker2, T. Ndungu1, D.E. Kaufmann2, V.O. Kasprowicz1 University of KwaZulu-Natal, Durban, South Africa; 2The Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA; 3 The Ragon Institute, Boston, MA, USA
1

Background: Due to the current dual-epidemic of HIV+/AIDS and TB, functional understanding of HIV+/MTB co-infection immunology is critical for furthering vaccine and therapeutic strategies against both pathogens. IL-10 is an anti-inflammatory cytokine, produced by lymphocytes and monocytes, that has been shown to down-regulate expression of anti-viral and antibacterial cytokines such as IFN-gamma, IL2 and TNF-alpha. Elevated IL-10 levels have been reported during chronic human HIV infection and ex vivo IL-10 receptor blockade in the same patients has been shown to increase HIV specific T cell function. Methods: This study assessed plasma levels of IL-10 and other cytokines (TNF, IFNy, IL-6, IL-2 and IL-13) in 119 subjects presenting different constellations of HIV and/or MTB monoor co-infection. Cytokine concentrations were measured by high sensitivity bead array assay (Luminex) and correlated with infection status, parameters of HIV disease progression and state of MTB infection. Furthermore, the effect of IL-10 receptor blockade (IL-10R) was assessed with regard to HIV and MTB specific CD4 T cell function in HIV+/LTBI co-infected subjects (as measured by Luminex). Results: Elevated levels of IL-10 were observed in HIV+/TB active individuals as compared to Active TB mono-infected(p=0.0295) and HIV mono-infected(p=0.0450). Plasma IL-10 levels were found to correlate directly with CD4 count (p=0.0002) and inversely with viral load (p=0.0168) in HIV+/LTBI subjects but not in HIV+/TB active subjects. Ex vivo blockade of IL-10R significantly enhanced MTB specific CD4+ T cell cytokine production as compared to isotype control in HIV+/MTB coinfected subjects: IFN-y (p=0.0078), TNF- (p=0.0078), IL-6 (p=0.0313) and IL-2 (p=0.0156). Conclusion: This study concluded that there is a synergistic upregulation of IL-10 in HIV+/TB active patients as compared to HIV and MTB mono-infection. Additionally, this study concludes that novel ex vivo enhancement of MTB and HIV specific CD4 T cell function is possible with IL-10R blockade in HIV+/LTBI subjects.

Universidad de Buenos Aires, Buenos Aires, Argentina; Hospital Juan A. Fernndez and Fundacin Huesped, Buenos Aires, Argentina; 3Fundacin Husped, Buenos Aires, Argentina; 4Sanatorio Otamendi, Buenos Aires, Argentina

Background: Understanding how immune homeostasis is perturbed during HIV infection is highly relevant. IL-17 is a protective cytokine against extracellular pathogens and helps maintain mucosal barrier. Objective: To evaluate IL-17-secreting T cell frequency during the first year following HIV Methods: Frozen PBMCs from 20 HIV+ subjects enrolled during seroconversion (by the Argentinean Group of Seroconverters), 12 HIV+ elite controllers (EC) and 9 healthy donors (HD) were used. Samples from seroconverters were obtained at enrollment (baseline), 3, 6, 9 and 12 months post-infection. Seroconverters were classified as Progressors if CD4 count dropped below 350 cells/microl or experienced AIDS-related B/C events within 12 months post-infection. IFN-gamma- and IL-17-secreting T cells were evaluated by flow cytometry upon stimulation with antiCD3/anti-CD28 antibodies or Results: CD3+CD4+: HIV+ subjects showed lower Th17 cell frequency compared to HD but higher Th17/Th1 ratio, indicating that IL-17-producing cells were enriched in the CD4+ subset. No difference between Progressor and NonProgressor seroconverters was observed at baseline samples. However, Progressors showed higher Th17 cell frequency and higher Th17/Th1 ratio than Non-Progressors (p=0.0049 and p=0.00485, respectively) at set-point. Neither Th17 cell frequency nor Th17/Th1 ratio changed significantly over time in both group of seroconverters. No significant difference was observed between seroconverters and EC. CD3+CD8+: Upon strong PBMC stimulation (PMA/Iono), EC showed higher frequencies of Tc17 cells, double-positive IL-17+IFN-+ cells and higher Tc17/Tc1 ratio than seroconverters (even at set-point samples) and HD (p<0.05 in all cases). Among seroconverters, no differences were observed between Non-Progressor and Progressors, independently of stimulation and time-point. However, Tc17 cell frequency significantly diminished between baseline and setpoint samples, regardless of progression status and stimulation. Conclusion: Results indicate that IL-17 produced by both CD4+ and CD8+ T cells might be an important mediator involved in host defense mechanisms against HIV-mediated immunophatogenesis during acute/early infection (CD4 and CD8) and aviremic chronic infection (CD8).

POSTERS 174 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.03


Preventing HIV-1 Infection, More Epitope Is Not Better: Analysis of Gag Epitope of 2 HLA Alleles Associated with Different Outcome of HIV Infection
M. Luo1, C. Semeniuk1, T. Diallo1, R. Capina1, J. Kimani2, C. Wachihi2, M. Kimani1, M. Mendoza1, S. Kiazyk1, T. Bielawny1, T.B. Ball1, F.A. Plummer1
1 2

Posters
P17.04
Epitope Mapping of HIV Clade A1-Specific CD8 T Cell Responses by Multiple Immunological Readouts Reveals Distinct Specicities Dened by Function
M. Richmond1, L.R. McKinnon2, S.A. Koester Kiazyk3, C. Wachihi4, M. Kimani4, J. Kimani4, F.A. Plummer3, T. Ball5 University of Manitoba, Winnipeg, Canada; 2University of Toronto, Toronto, Canada; 3National Microbiology Laboratory, Winnipeg, Canada; 4University of Nairobi, Nairobi, Kenya; 5National HIV and Retrovirology Laboratories, Winnipeg, Canada
1

National Microbiology Laboratory, Winnipeg, Canada; University of Nairobi, Nairobi, Kenya

Background: Current HIV vaccine candidates have been based on the conventional views of viral infection and attempt to induce broad T cell responses to HIV-1. Until now, the candidate vaccines based on such approach either failed to provide protection or produced modest effect that is not satisfactory for an effective vaccine. Since these vaccine candidates were not based on the correlates of protection against HIV-1 infection, improving such understanding could be critical for successful vaccine development. Methods: A subset of women enrolled in the Pumwani Sexworker Cohort remain uninfected by HIV-1 despite repeated exposures through sex work. This resistance to HIV-1 infection is associated with several alleles of Human Leukocyte Antigens (HLAs) and specific CD8+ and CD4+ T cell responses. In this study we systematically analyzed HIV-1 clade A and D Gag epitope profiles of two HLA class I alleles associated with different outcomes of HIV-1 infection, A*0101 is significantly associated with slower seroconversion while B*0702 is associated with rapid seroconversion. We screened a Gag peptide library with iTopia Epitope Discovery System to compare the peptide binding capacity of these two alleles. The identified peptides were characterized by affinity and off-rate assays and confirmed by interferon gamma ELISPOT assays using patient peripheral blood mononuclear cells. Results: A*0101, an allele associated with protection from HIV1 infection, only binds to 3 epitopes in Gag. Whereas, B*0702, an allele associated with rapid disease progression, has 30 Gag epitopes. There is no significant difference in peptide binding affinity, off-rate, ELISPOT SFU values and epitope specific Tem/ Tcm frequencies. Conclusion: In contrast to the broad peptide binding spectrum of B*0702, A*0101s epitopes are narrowly directed. Observations of this study question the current approach for HIV-1 vaccine development and propose a different vaccine development strategy.

Background: The limited success of HIV vaccine candidates to date highlights our need to better characterize protective cellmediated immunity. While HIV-specific CD8+ T cell responses have been largely defined by measuring IFN-, these responses are not always protective, and it is unclear whether the same epitopes would predominate if other functional parameters were examined. Methods: Previously, we measured CD8+ T cell responses to clade A1 HIV p24, in a Kenyan commensal sex worker cohort, using an unbiased epitope mapping approach and identified 50 functionally specific-epitopes. Here we extended our findings and further characterize HIV-specific CD8+ T cell responses measuring seven CD8+ T cell functions (IFN-, CD107a, MIP-1a, MIP-1, TNF-, IL-2 and proliferative capacity) in 80 chronically HIV-infected individuals to eleven identified epitopes. Results: Epitope mapping revealed that most epitope-specific responses were IFN- negative (50/69). Many responses had polyfunctional (33%) and proliferative (19%) components. An inverse association between IL-2 and proliferation responses was also observed, contrary to what has been described. Characterization of the eleven epitopes of interest was consistent with our previous findings. Most epitope-specific responses were IFN- negative (64%) and many responses were polyfunctional (38%). Two of the eleven epitopes were recognized at significantly higher frequencies (p<0.02) and we identified epitopes that preferentially elicited specific cytokines (p<0.02). Preliminary data suggests that some of these epitopes are significantly more likely to elicit a polyfunctional response. Conclusion: Together, these data suggest that the specificity of CD8+ T cell responses differs depending on immunologic readout, with a 3.5-fold increase in breadth detected by including multiple parameters. Furthermore, identification of epitopes that elicit polyfunctional responses reinforces the need for the comprehensive evaluation of HIV vaccine candidates, and may represent novel targets for CMI-based vaccines.

AIDS Vaccine 2011

175

POSTERS

Posters
P17.05

Topic 17: T Cell Immunity P17.06


Cryptic Epitopes from Alternative Reading Frames Restricted by HLAs Associated with a Good Prognosis Are Frequently Recognized in HIV Infection
A. Bansal1, S. Seevers1, T. Mann1, J. Carlson2, A. Bet1, Q. Zhao1, S. Heath1, D. Heckerman2, P.A. Goepfert1
1 2

Characterization of CD8+ T Cell Epitopes Recognizing HIV-1 Epitope Variants


M.M. Herman1, L.R. McKinnon2, S. Kiazyk3, J. Kimani4, T. Ball3, F.A. Plummer3 University of Manitoba, Winnipeg, Canada; 2University of Toronto, Toronto, Canada; 3Public Health Agency of Canada, Winnipeg, Canada; 4University of Nairobi Institute of Tropical and Infectious Diseases, Nairobi, Kenya
1

University of Alabama at Birmingham, Birmingham, AL, USA; Microsoft Corporation, Seattle, WA, USA

Background: One major challenge in the creation of an HIV-1 vaccine is the extreme genetic diversity of the virus. It is thought that a more cross-reactive T-cell response would be beneficial in circumventing this issue. Despite this, little is known about the characteristics of these responses in nature, and many aspects of the variant-epitope CD8+ T-cell response remain poorly defined. Here, we characterize CD8+ T-cells specific to an immunodominant HIV-1 epitope, IW9, and 2 of its variants, to better understand the level of cross-reactivity between them. Methods: Using samples from the Pumwani commercial sexworker cohort in Nairobi, Kenya, individuals positive for either HLA-B*4201 or HLA-B*0702 were screened for binding to tetramers specific for the IW9 epitope and 2 variants. Analysis of epitope specific cytokine expression and proliferation was performed to determine the level of functional cross-reactivity of the variantspecific T-cell pools. Additionally, heteroduplex mobility assays (HDMAs) were performed to determine differences in TCR usage among the T-cells responding to each variant. Results: Tetramer co-staining experiments indicate that there is some cross-reactivity among these variants, shown by strong double-positive populations in flow cytometry. However, in some cases, separate cell populations are being recognized by each variant. Proliferation assays show that T-cells stimulated by one variant can be recognized by tetramers specific to another variant. HDMAs revealed that there are differences in TCR usage among variant-specific CD8+ T-cells both at the family and clonotype level. This may suggest a combination of public and private clonotype usage, which could explain the differences in cross-reactivity and functionality. Conclusion: In order to create a T-cell vaccine that can target and produce protective immune responses, it will be necessary to fully understand the nature of cross-reactive responses. This study has shown evidence that cross reactivity exists within the IW9 epitope, and that there are several factors that may affect this.

Background: Cryptic epitopes (CE) are products of translation of alternate reading frames (ARF, 2 sense and 3 antisense) that are commonly targeted during HIV/SIV infection. To understand the full extent of contributions of HIV specific CE in HIV-1 pathogenesis, we performed mapping of the CE derived from ARFs of nine HIV-1 encoded proteins. Methods: For a comprehensive approach, we designed overlapping peptides (OLPs; 12-18 mers) for the 5 ARFs of clade B gag and pol (PeptGen program, Los Alamos). We also predicted (using clade B sequences/Epipred program) 9-11mer peptides that bind HLA-B*27, B*57 or B*5801 and span the ARFs of the nine HIV-1 encoded proteins. PBMC from seronegative donors (n=42) and chronically (CHI, n=93) HIV clade B infected patients were used to evaluate CE specific T-cell responses in an IFN-g ELISpot assay. Among CHI, 80% patients had either B*27, B*57 and/or B*5801 alleles. Results: We predicted 520 (B*27), 955 (B*57) and 1027 (B*5801) potential CE in each of the five ARFs of the nine HIV-1 encoded proteins. The antisense frames encoded a majority of these CE. Predicted peptides with >50% probability of being an epitope were synthesized; B*27 (N=30), B*57 (N=39) and B*58 (N=90). Overall, 26% and 2% of CHI patients and seronegative donors respectively had CE responses (p=0.0006, Fischers exact). The overall responder frequency for HLA specific CE response was 22% (range 6%-43%) with the highest recognition frequency noted for HLA-B*5801/B*57 patients (p=0.005, Fischers exact). In our comprehensive mapping analysis, 17% patients targeted CE from gag/pol ARFs and all responders were either HLA-B*27, B*57 or B*5801. Among these, the most frequently targeted were pol 2 (31%) followed by gag 2 and 5 (25%) frames. Conclusion: These data underscore the importance of CE targeting especially those that are presented by the so-called protective alleles in HIV-1 infection and hence have implications for vaccine design.

POSTERS 176 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.07


Differences in Magnitude, Breadth and Targets of HIV-Specific Response in Progressive Versus NonProgressive Acute/Early HIV Infection
G. Turk1, Y. Ghiglione1, M.E. Socas2, A.M. Rodriguez1, J. Favilene1, M.J. Ruiz1, O. Sued3, D. Pryluka4, N. Laufer1, P. Cahn5, H. Salomon1, M.M. Gherardi1
1

Posters
P17.08
HIV-1-Specific Cytotoxic T Lymphocytes CrossRecognizing an Escape Mutation at Early Phase of HIV-1 Infection
T. Akahoshi1, H. Gatanaga2, S. Oka2, M. Takiguchi1
1

Centro Nacional de Referencia para el SIDA, Buenos Aires, Argentina; 2Hospital Juan A. Fernndez and Fundacin Huesped, Buenos Aires, Argentina; 3Fundacion Huesped, Argentina; 4Sanatorio Otamendi, Buenos Aires, Argentina; 5 Hospital Juan A. Fernndez and Fundacion Huesped, Buenos Aires, Argentina
Background: Characterizing HIV-specific immune response during acute infection and how it is modulated over time is highly relevant at HIV-vaccine design setting. Objective: To study HIV-specific T cell responses in Argentinean subjects (enrolled by the Argentinean Group of Seroconverters), during HIV seroconversion and up to the first year of infection Methods: Frozen PBMCs from 21 HIV+ seroconverters and 10 HIV+ elite controllers (EC) were used. Samples from seroconverters were obtained at enrollment (baseline), 3, 6 and 12 months post-infection. Seroconverters were classified as Progressors if CD4 count dropped below 350 cells/microl or experienced AIDS-related B/C events within the first year postinfection. HIV-specific T cell responses were evaluated by IFNgamma ELISPOT. Magnitude, breadth and spot size (as indicator of released IFN-gamma) were compared inter- and intra-groups at baseline and follow-up samples, using parametric and nonparametric statistics Results: Magnitude: Among seroconverters, Gag was preferentially targeted in Non-Progressors (NP) and Nef in Progressors (P). In EC, responses against Gag were predominantly found (Gag vs Nef and Env p<0.005). Within Gag, p24-specific cells dominated in NP and EC while p17 and p2-p7-p1-p6-specific cells dominated in P. Both at baseline and set-point samples, Gagspecific responses versus viral load and Nef-specific responses versus CD4 counts correlated significantly (inversely and directly, respectively). Breadth: Both at baseline (p=0.0115) and set-point samples (p=0.0428), P recognized fewer viral regions than EC. Spot size: NP and EC produced larger spots than P both when stimulating cells with HIV-specific peptides (p=0.0017 and p=0.0002, respectively) or CEF (CMV, Epstein Barr, Influenza)specific peptides (p=0.02 and p=0.0039, respectively). This trend was maintained over time Conclusion: Early T cell responses (at baseline and set-point samples) preferentially targeting p24, higher breadth and larger spot size were associated with non-progressive acute/early infection indicating that these factors play an important role in early viral containment and should be considered for rational vaccine design

Center for AIDS Research, Kumamoto University, Kumamoto, Japan; 2AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan

Background: Previous studies showed that HIV-1-specific cytotoxic T lymphocytes CTLs recognizing both wild-type and escape-mutant epitopes (cross-reactive CTLs) exist in vivo. However, a role of cross-reactive CTLs in the control of HIV-1 replication remains unclear. We here studied cross-reactive CTLs specific to the HLA-A*2402-restricted Gag28-36 (KYKLKHIVW: WT) epitope and its escape mutant (3R). Methods: We analyzed 11 HLA-A*2402+ individuals with primary HIV-1 infection who were monitored from early to chronic phases. Results: Sequence analysis of plasma viruses at an early phase showed that three and eight patients were infected with WTand 3R virus, respectively. Analysis of bulk CTLs stimulated with the WT or 3R peptides showed that cross-reactive CTLs were elicited at an early phase in three (KI-092, -158, and -161) and one (KI091) of the eight patients. WT and 3R-specific CTLs were also detected in two (KI-092 and -161) and four (KI-134, -136, -151, and -163) patients, respectively. 3R peptide-induced CTL clones established from KI-091 (KI-091 CTL clones), which recognized the WT and 3R peptides equally, lysed both WT and 3R virus-infected cells. In contrast, WT peptide-induced CTL clones established from KI-092 (KI-092 CTL clones), which recognized the WT peptide more effectively than 3R, lysed only WT virus-infected cells. However, in vitro replication suppression assay showed that the KI-091 CTL clones failed to suppress the replication of both viruses, whereas the KI092 CTL clones effectively suppressed that of the WT virus. Conclusion: These results show that cross-reactive CTLs are elicited at an early phase in some individuals infected with the WT virus before the selection of the 3R one or with the 3R virus and that their ability to suppress the replication of HIV-1 is much lower than that of WT-specific CTLs. The present study suggests that cross-reactive CTLs can not effectively control the WT and 3R-escape-mutant viruses.

AIDS Vaccine 2011

177

POSTERS

Posters
P17.09

Topic 17: T Cell Immunity P17.10


Virus Suppressive Activities of CD8+ T Cells Correlate with SIV-Specific CD8+ T Cell Responses
H.S. Hong1, J. Gillis1, Y. Yu1, F.E. Wong1, R.P. Johnson1
1

Interdisciplinary Evaluation of Broadly-Reactive HLA Class II Restricted Epitopes Eliciting HIVSpecific CD4+ T Cell Responses
M. Buggert , M. Norstrm , C. Lundegaard , O. Lund , M. Nielsen2, A.C. Karlsson1
1 1 2 2 1

Harvard Medical School, Southborough, MA, USA

Karolinska Institutet, Stockholm, Sweden; 2Technical University of Denmark, Department of Systems Biology, Denmark

Background: CD4+ T cells orchestrate immune protection by helping other cells of our immune system to clear viral infections. It is well known that the preferential infection and depletion of CD4+ T cells contributes to hampered systemic T cell help following HIV infection. However, the functional and immunodominant discrepancies of CD4+ T cell responses targeting promiscuous MHC II restricted HIV epitopes remains poorly defined. Thus, utilization of interdisciplinary approaches might aid revealing broadly-reactive peptides eliciting CD4+ T cell responses. Methods: We utilized the novel bioinformatic prediction program NetMHCIIpan to select 64 optimized MHC II restricted epitopes located in the HIV Gag, Pol, Env, Nef and Tat regions. The epitopes were selected to cover the global diversity of the virus (multiple subtypes) and the human immune system (diverse MHC II types). Optimized polychromatic flow cytometry analysis, including the functional markers IFN, IL-2, IL-21, MIP-I and TNF, revealed immunogenicity of the individual epitopes. The study subjects (n=38) were of diverse ethnical background infected by different HIV subtypes. High resolution HLA typing and sequences of the HIV-Gag and Nef regions were obtained. Results: The FACS analysis revealed immunogenicity against 73% of the epitopes. All subjects, except one, recognized at least one epitope. Interestingly, almost all epitopes located in Gag (15/15) and Nef (14/15) elicited responses, while epitopes in Pol (10/15) and Env (5/15) revealed restricted CD4+ T cell immunogenicity. This difference in immunogenicsity between the regions was significant (One-way ANOVA: p < 0.001). Additionally, Gag and Nef epitopes generated greater polyfunctionality than Pol- and Env-specific CD4+ T cells. Importantly, we found that the use of optimized epitopes improved the polyfunctionality compared with overlapping HIV Gag (p55) peptides. Conclusion: Using an unbiased approach where we have predicted peptides with same prerequisites, we demonstrate that HIV-specific CD4+ T cell immunodominance is heavily skewed, targeting particularly Gag and Nef.

Background: The immune correlates of the remarkable efficacy of protection induced by vaccination of rhesus macaques with SIVnef remain elusive. We addressed the question whether CD8+ T cells from SIVnef-vaccinated animals could suppress SIV replication in autologous CD4+ T cells and whether the virus suppressive activity correlated with SIV-specific immune responses. Methods: CD4+ T cells from 5 uninfected, 13 WT-SIV-infected and 19 SIVnef-vaccinated animals were infected with SIVmac239 and co-cultured with purified CD8+ T cells at a wide range of effector-to-target- (E:T) ratios. Intracellular expression of p27 was measured and IC50-values denoting the E:T ratio providing 50% inhibition were determined. ELISPOT assays and intracellular cytokine assays were performed. Statistical analysis included Mann-Whitney test, One-Way ANOVA followed by Tukey test and Pearson correlation analysis. Results: Determining IC50-values significantly improved the dynamic range as revealed by substantial differences in virus suppressive activity (>38-fold) in either WT-SIV or SIVnefinfected animals compared with uninfected animals (P < 0.0001). No differences in virus suppressive activity were observed between SIVnef-vaccinated and WT-SIV-infected animals. The magnitude of immune responses as detected by IFN- ELISPOT assays did not differ between WT-SIV- and SIVnef-infected animals, nor did ELISPOT responses correlate with the potency of virus suppression by CD8+ T cells. Intracellular cytokine staining assays revealed similar levels of IFN-, TNF-, IL-2, MIP-1 and CD107a responses following Gag- or Env stimulation in SIVnefvaccinated and WT-SIV-infected animals. However, we identified significant correlations of Gag-specific IFN--, TNF--, MIP1- and CD107a- CD8+ T cell responses with virus suppressive activity. Conclusion: CD8+ T cells from SIVnef-vaccinated animals are able to mediate potent inhibition of SIV replication in autologous CD4+ T cells, with activity that was similar to that observed in WT-SIV-infected animals. The magnitude of virus suppression correlated with the magnitude of SIV-specific CD8+ T cell responses, suggesting that these properties are closely linked.

POSTERS 178 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.11


High TCR Avidity HIV-Specific CD8 T-Cells Accumulate Over Time During HIV Infection and Are Associated to High PD1 Expression
S. Vigano1, A. Savoye1, M. Perreau1, F. Bellutti Enders1, P. Bart1, G. Pantaleo1, A. Harari1
1

Posters
P17.12
Spontaneous HIV-1 Control Is Associated with Distinct Epitopes that Promiscuously Bind Multiple HLA Class II Alleles
S. Ranasinghe1, M. Flanders1, D. Soghoian1, S. Cutler1, M. Lindqvist1, I. Davis1, M. Ghebremichael1, B.D. Walker1, A. Sette2, F. Pereyra1, D. Heckerman3, H. Streeck1 Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2La Jolla Institute for Allergy and Immunology, Center for Infectious Dis, La Jolla, CA, USA; 3Microsoft Research, Los Angeles, CA, USA
1

Laborator of AIDS Immunopathogenesis, Service of Immunology and Allergy, Lausanne, Switzerland

Background: CD8 T-cells play a critical role in antiviral immunity. However, mechanisms of virus control and immune correlates of protection are still not fully understood. Among other factors, TCR avidity (i.e. antigen sensitivity) is thought to play a critical role. High TCR avidity T-cell responses are associated to virus clearance; however, their relevance in chronic persistent viral infections remains unclear. Methods: 183 HIV-, CMV-, EBV- and Flu-specific CD8 T-cell responses were analyzed in 61 patients with early acute or chronic (progressive and non-progressive) HIV infection and in 32 HIV-negative subjects. TCR avidity (measured by the effect concentration 50% [EC50]) as well as T-cell exhaustion/ senescence/differentiation and functional profile were determined. Results: TCR avidity of Flu- (acute virus) and HIV (acute infection)specific CD8 T-cells was significantly lower than those of chronic CMV/EBV- and HIV (progressive and nonprogressive chronic infection)-specific CD8 T-cells (P<0.01). TCR avidity remained higher in HIV-specific CD8 T-cells from chronic as compared to acute patients when analyses were restricted to common peptideHLA relationships (P<0.0003) and high avidity responses were associated to lower expression of co-stimulatory molecules (CD27, CD28 and CD134; all P<0.0001) and to a more differentiated/ effector phenotype (P<0.001). Furthermore, TCR avidity increased overtime in untreated acute or chronic infection (P<0.01) but not in successfully treated acute or chronic infection (P>0.05) paralleled by a significant loss of polyfunctionality and cytotoxic potential. Consistently, high TCR avidity HIV-specific CD8 T-cell responses were also correlated to high PD1 expression (P<0.04. Conclusion: These results suggest that high TCR avidity HIVspecific CD8 T-cell responses accumulate during the natural course of HIV infection and that continuous exposure to HIV-1 antigens is a potential driving mechanism which also leads to functional anergy and T-cell exhaustion. These results advance our understanding of the relationship between TCR avidity, Ag exposure and T-cell exhaustion of antiviral CD8 T-cell responses.

Background: Although HIV-specific CD4+ T-cells are preferentially infected, there is growing evidence that these cells play a pivotal role in control of viremia. However, little is known about the recognition and HLA-class II restriction of HIVspecific CD4+ T-cells in the setting of chronic or spontaneously controlled HIV-infection. Yet, this knowledge will be crucial for the induction of protective immunity in a prophylactic vaccine. Methods: CD8+ depleted PBMCs from 94 HIV-infected subjects (controller, progressor and ART-treated) were screened for IFNresponses to 410 overlapping clade B peptides in a modified Elispot. HLA-class II restriction was defined by testing CD4+ T-cell lines against peptide-loaded L-cells transfected with a single HLA-DR. Results: HIV-specific CD4+ T-cell responses were detected in all patient subgroup yet the breadth of these responses was significantly expanded in HIV-controllers (p=0.029). Most HIV-specific CD4+ T-cells targeted epitopes within Gag, Nef and gp120. Strikingly, we observed significant differences in the immunodominance profile between patient subgroups that distinguished not only elite controllers from rapid progressors, but also from viremic controllers. While elite controllers dominantly targeted a tight cluster of conserved epitopes within p24, chronic progressors preferentially targeted epitopes within the C1/C3 domain of gp120. Moreover, the ratio of Env- and Gag-specific responses was a clear indicator of viral control. A multivariate bootstrap analysis identified four distinct Gag epitopes that were associated with spontaneous control (Probability=0.60-0.85), while a single gp120 peptide in C3 was associated with high viremia (Probability=0.82). Detailed characterization showed that promiscuous binding to multiple HLA-DR alleles occurs frequently, and revealed that HLA-DRB1*0701 is associated with slow disease progression (Probability=0.88). Conclusion: Our data demonstrate that significant differences exist in protein targeting by HIV-specific CD4+ T-cells between controllers and progressors. We also identified distinct epitopes associated with viral control that are conserved and bind promiscuously to multiple HLA-class II alleles, which will be important for vaccine design.

AIDS Vaccine 2011

179

POSTERS

Posters
P17.13

Topic 17: T Cell Immunity P17.14


In Vitro CD8-Mediated Inhibition of SIV Correlates with In Vivo Virologic Control in Rhesus Macaques
K.E. Stephenson1, D.H. Barouch1
1

Role of Hexon- and Fiber-Specific Antibodies in Ad5 Neutralization


R.R. Bradley1, D.H. Barouch1
1

Beth Israel Deaconess Medical Center, Boston, MA, USA

Beth Israel Deaconess Medical Center, Boston, MA, USA

Background: The immunogenicity of adenovirus serotype 5 (Ad5) vectors has been shown to be suppressed by neutralizing antibodies (NAbs) against the hexon hypervariable regions (HVRs). The role of NAbs directed against the fiber, however, remains unclear. Methods: Chimeric recombinant Ad5 vectors containing the hexon HVRs of Ad48 (Ad5HVR48), the fiber knob of chimpanzee AdC68 (Ad5KC68), or both (Ad5HVR48KC68) were constructed. All vectors exhibited comparable growth and infectivity parameters. Human and murine sera were assessed for NAb responses against each vector. C57BL/6 mice, either nave or preimmunized twice with 1010 vp of Ad5-Empty, were immunized with 109 viral particles (vp) of each vector expressing SIV Gag. Gag-specific immune responses were assessed by tetramer, ELISPOT, and ICS assays. Results: Serology studies in both Ad5-preimmunized mice and 116 healthy Ad5-seropositive humans from sub-Saharan Africa demonstrated that Ad5-specific NAbs were directed primarily against the hexon HVRs. Residual non-hexon NAbs were largely directed against the fiber knob. Ad5, Ad5KC68, Ad5HVR48 and Ad5HVR48KC68 vectors expressing SIV Gag proved comparably immunogenic in nave mice by tetramer, ELISPOT, and ICS assays. In the presence of Ad5-specific preexisting immunity, however, the immunogenicity of Ad5 and Ad5KC68 was abrogated, and only Ad5HVR48 and Ad5HVR48KC68 were immunogenic. Ad5HVR48KC68 evaded preexisting Ad5 immunity more effectively than did Ad5HVR48. Conclusion: These data indicate that the hexon HVRs are the primary target of Ad5 NAbs following both Ad5 vaccination and natural Ad5 exposure. Residual non-hexon NAbs are directed largely against the fiber knob. Exchanging both the hexon HVRs and the fiber knob results in a vector that nearly completely evades Ad5-specific NAbs.

Background: The in vitro viral inhibition assay was developed to measure CD8-mediated control of HIV-1 replication. However, the utility of the in vitro SIV inhibition assay remains to be determined. We therefore optimized the SIV inhibition assay in a cohort of vaccinated and SIV-infected rhesus macaques to explore correlates of virologic control. Methods: PBMC were isolated from 10 rhesus macaques infected with SIVmac251 with viral loads ranging from 3.2 to 6.9 log10 SIV RNA. All animals had been previously vaccinated with MVA/Ad26, Ad26/MVA, or a sham vaccine. CD8-depleted PBMC were cultured with and without CD8 cells for 2 weeks, and p27 content was measured by ELISA on days 7 and 14. CD8-mediated inhibition was expressed as the log10 reduction in p27 content between cultures of mixed CD8 and CD8-depleted cells, compared to CD8-depleted cells alone. Results: CD8-mediated inhibition on day 7 ranged from 0 to 2.1 log10 reduction in p27, with median inhibition of 0.7 (SD 0.57). In vitro inhibition was significantly inversely correlated with plasma SIV RNA (p=0.0104) with a Pearson correlation coefficient of -0.76 (CI -0.94 to -0.25). Inhibition ranged from 0 to 2.8 on day 14, with median inhibition of 0.9 (SD 0.9). The correlation between in vitro inhibition and plasma SIV RNA trended towards significance (p=0.0974) on day 14 but was weaker than on day 7. Conclusion: CD8-mediated inhibition of SIV in vitro correlates with virologic control in vivo. The performance of this assay was optimal when measuring endogenous SIV replication after 7 days of culture. These data suggest that this viral inhibition assay will be useful for evaluating immune correlates of protection in nonhuman primates.

POSTERS 180 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.15


Accurate Recapitulation of Human HIV-Specific CD8+ T Cell Responses in BLT Humanized Mice During Acute Phase Infection
T.E. Dudek1, D.C. No1, E. Seung2, V. Vrbanac2, L. Fadda1, P. Bhoumik1, A.D. Luster3, M. Altfeld1, A.M. Tager2, T.M. Allen1 Ragon Institute of MGH, MIT and Harvard, Charlestown, MA, USA; 2Center for Immunology and Inflammatory Diseases, Massachusetts General, Boston, MA, USA; 3Center for Immunology and Inflammatory Diseases, Massachusetts Genera, Boston, MA, USA
1

Posters
P17.16
The CD8+ T Cell Antiviral Inhibitory Activity Predicts the Rate of CD4+ Cell Decline in Early HIV-1 Infection and Is Independent of Viral Subtype
H. Yang1, H. Wu2, S.A. Freel3, H. Yan2, X. Huang2, X. Xu1, J. Birks4, G.D. Tomaras3, A. McMichael1, L. Dorrell1
1

Background: The SIV-infected macaque has proven to be an extremely valuable model of HIV infection. However, differences in host genetics between humans and macaques, and substantial sequence differences between HIV and SIV, limit its ability to accurately model HIV-specific human immune responses. While humanized mice have the potential to fill this void, little data exists regarding whether their pathogen-specific responses accurately recapitulate those observed in human infections. Methods: Human liver, thymus and CD34+ hematopoietic stem cells derived from 6 distinct human donor tissues encoding unique HLA haplotypes were used to generate 38 humanized BLT mice. Genome-wide HIV-1 sequence evolution was used as an initial marker of early HIV-specific CD8+ T-cell responses. IFNgamma ELISpot and intracellular cytokine staining (ICS) assays were employed to identify antigen specific CD8 responses. Results: Following HIV infection, peak viral loads and set points above 1.0x105 copies/ml were observed. Mice expressing HLA-A01 and Cw03 exhibited reproducible and rapid viral escape within two CD8 epitopes in Env and Nef, respectively. Two other groups of mice expressing either A01 or Cw03 revealed similar evolution. These epitopes are immunodominantly targeted during natural acute HIV infection of humans, and IFN-gamma ELISpot and ICS assays confirmed the presence of CD8 responses to these epitopes. CD8 responses against the normally acute phase HLA-B57 restricted TW10, IW9 and KF11 epitopes in Gag were detected in mice expressing HLA-B57 at ex-vivo frequencies similar to those seen in humans. The presence of the HLA-B57 allele was also associated with a statistically significant reduction in viral loads between 6-12 weeks post infection. Conclusion: The specificity, magnitude and immunodominance patterns of HIV-specific CD8 responses in humanized BLT mice appear to accurately reflect those of HIV-infected subjects, supporting the potential of this small animal model to explore the efficacy of natural and vaccine-elicited cellular immune responses.

Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom (Great Britain); 2YouAn Hospital, Capital Medical University, Beijing, China; 3Duke University Human Vaccine Institute and Department of Surgery, Durham, NC, USA; 4Centre for Statistics in Medicine, University of Oxford, Oxford, United Kingdom (Great Britain)

Background: The lack of a reliable marker of protective immunity and the genetic diversity of HIV-1 are major obstacles to the development of an effective HIV-1 vaccine. Rare individuals who control HIV-1 without treatment show potent CD8+ T cell-mediated viral inhibition in vitro, indicative of effective immune control. We investigated the relationship between CD8+ T cell antiviral activity and the rate of CD4+ cell loss in early HIV-1 infection. Methods: We studied individuals with documented recent infection to determine the capacity of ex vivo CD8+ T cells to suppress replication of an exogenous HIV-1 isolate. We quantified HIV1BaL super-infected CD4+ T cells by intracellular p24 staining, after culture in the presence or absence of autologous CD8+ T cells. We studied prospectively the interaction between CD8+ T cell antiviral activity and the rate of CD4+ cell decline during followup. We also evaluated the ability of CD8+ T cells to inhibit 5 HIV-1 isolates representing different clades in 15 HIV-1 controllers. Results: In 20 recently infected subjects CD8+ T cell antiviral activity against a single heterologous virus was strongly predictive of the rate of subsequent CD4+ cell decline during 3 years follow-up (p <0.0001). This interaction was independent of patients viral subtype (CRF01_AE in 45%, B / CRF07_BC in 50%). In a separate analysis of HIV-1 controllers, potent crossclade CD8+ T cell inhibition was observed; however, there was no clear relationship between suppression of HIV-1BaL and other HIV-1 isolates. Conclusion: The antiviral inhibitory capacity of CD8+ T cells is strongly predictive of the rate of CD4+ cell decline in early HIV-1 infection, irrespective of the infecting viral subtype. This CD8+ T cell antiviral inhibition assay has potential as a tool to evaluate the potency of HIV-1 vaccine candidates. Preliminary data suggest that CD8+ T cells, which can potently inhibit HIV1BaL show cross-clade suppressive activity.

AIDS Vaccine 2011

181

POSTERS

Posters
P17.17

Topic 17: T Cell Immunity P17.18


Up-Regulation of LAG-3 Expression on T Cells in HIV-1 Infection Is Correlated with Disease Progression
X. Tian1, A. Zhang1, C. Qiu1, S. Yuan1, S. Qiu1, H. Hu1, W. Wang1, C. Qiu1, X. Zhang1, J. Xu1
1

Prevalence, Incidence, Risk Factors and Willingness to Participate in HIV Vaccine Trials Among Gay and Bisexual Men and Transgender Persons Seeking HI
J. Xu , A. Zhang , W. Wang , M. Zhou , C. Qiu , S. Yuan , S. Qiu1, H. Hu1, J. Xiao1, C. Qiu1, X. Zhang1, J. Xu1
1 1 1 1 1 1 1

Shanghai Public Health Clinical Center, Shanghai, China

Institutes of Biomedical Sciences,Fudan University, Shanghai, China

Background: The role of CD160 in CD4 T and NK cells has been intensively explored, whereas its role in CD8+ T cells remains poorly understood Methods: We used multi-color flow cytometry to characterized the co-expression of CD160 with PD-1, CD38, HLA-Dr, CCR7, CD45RA and CD62L on circulating CD8+ T cells derived from cohorts of HIV-1 sero-negative and HIV-1 infected subjects including slow progressors (SP) and long term non-progressors (LTNPs) who maintained stable CD4 counts over 400 and low HIV-1 viral loads in the absence of HARRT for more than 10 years. Furthermore we have investigated the ex vivo function of HIV/CEF-specific CD160+ CTL by multiple assays including IFNgamma/Granzyme B based ELISPOT, intracellular Granzyme B/ Perforin staining and CD107a degranulation etc. Results: We showed that the expression of CD160 on CD8+ T cells was significantly up-regulated during HIV-1 infections. The increased percentage of CD160+ CD8+ T cells in TEM/TEMRA cells during HIV infection, which was significantly higher in LTNP than in SP, was associated positively with CD4 counts but inversely with HIV-1 viral loads. CD160+ CTLs exhibited a dominant PD-1- CD38- HLA-DR+ CCR7- CD62L- phenotype. The intracellular staining data of non-stimulated PBMC showed that the proportions of CD160+ Granzyme B+ / Perforin+ CTL in HIV-1 LTNPs are significantly higher than CD160- Granzyme B+ / Perforin+ CTL. Furthermore, we observed that stimulation with anti-CD160 antibody resulted in increased HIV-gag specific Granzyme B but not IFN-gamma ELISPOT responses. We also found that HIV-gag/CEF-specific degranulation ability of CD160+ CTL was stronger than that of CD160- CTL, addition of anti-CD160 antibody led to significantly enhanced degranulation capacity of CD160high CTL subsets. Conclusion: Our data indicates that engagement of CD160 may trigger a co-stimulatory signal pathway for cytotoxic function of CD8+ T cells and thereby play an immune protective role in HIV-1 infection.

Background: Lymphocyte activation gene-3 (LAG-3), expressed on activated CD4+ and CD8+ T cells, is known to negatively regulates T-cell responses, but its role in HIV-1 infection in vivo remains unclear. Methods: In the present study, we analyzed LAG-3 expression differences between HIV-uninfected and HIV-infected individuals using microarrays, quantitative PCR and flow cytometry. Results: We found that LAG-3 expression in PBMC cells was higher in HIV-1 infected individuals than HIV sero-negative persons. LAG-3 expression was correlated with disease progression: LAG3 expression was up-regulated on both CD4 and CD8 T cells from HIV-1 infected individuals, which was also correlated positively with viral load and inversely with CD4 count. We further characterized Lag-3+CD4 and Lag3+CD8 T cells. The coexpression of LAG-3 and immune activation marker was analysis, indicating that LAG-3 is an important active hallmark of T cell. The co-expression of Lag-3 and an immune inhibitory receptor (PD-1) and a prototypical death receptor (CD95) is low, indicating that Lag-3 and inhibitory receptor PD-1 are expressing on different subsets of T cells and have not obvious role in promoting apoptosis of CD8 T cells. We showed that HIV-specific Lag-3 expression is not recovered by HAART. Our experiments also find that Lag-3 primarily expresses on terminal memory effector CD8+T cells irrespective of HIV- or HIV+. Conclusion: In conclusionLag-3 is up-regulated concurrently with HIV-1mediated chronic immune activation in comparison with HIV-1 sera-negative controls and may represent a novel target for the therapeutic reversal of HIV-1 associated T cell dysfunction. In addition, blockade of Lag-3 enhanced cytokine production such as TNF-, IL-2 and IFN- while lowering cytokine production, such as IL-4 and IL-17.

POSTERS 182 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.19


Low Level of Regulatory T Ccells and Maintenance of Balance Between Regulatory T Ccells and TH17 Ccells in HIV-1+ Elite Controllers
L. Brandt1, T. Benfield2, H. Mens2, L.N. Clausen2, T.L. Katzenstein3, I. Karlsson1 Statens Serum Institut, Copenhagen S, Denmark; 2Department of Infectious Diseases, Hvidovre University Hospital, Denmark; 3 Department of Infectious Diseases, Copenhagen University Hospital, Denmark
1

Posters
P17.20
Contribution of Magnitude and Multifunctional HIV-Specific CD8+ T Cells to Immune Selective Pressure
C. Riou1, M. Mlotshwa2, M. Abrahams1, S. Goodier1, F. Treurnicht1, K. Mlisana3, S. Abdool Karim 3, C. Williamson1, C.M. Gray1, T. CAPRISA 002 study team3
1

Institute of Infectious Diseases and Molecu, University of Cape Town, Cape Town, South Africa; 2National Institute for Communicable Diseases, Johannesburg, South Africa; 3 CAPRISA, University of KwaZulu Natal, Durban, South Africa
Background: Defining functions of CD8+ T cells that provide immune selective pressure is important for understanding potential vaccine-induced responses. We aimed to identify functional profiles of CD8+ T cells associating with epitope escape. Methods: For each studied individual (n=5), autologous epitopes, that underwent early and late mutation after infection, were identified through SGA. We used polychromatic ICS to measure CD8+ T cell responses to peptides corresponding to: early variant epitopes (EV, autologous transmitted sequences prior to early mutation); mutant epitopes (ME, autologous sequences after mutation); and late variant epitopes (LV, autologous transmitted sequences undergoing late mutation). Magnitude, function (CD107, IFNg, TNFa, MIP1b, Perforin) and memory maturation (CD45RO, CD27) were assessed longitudinally. Results: Prior to epitope mutation, early variant epitope (EV)specific CD8+ responses were significantly (p=0.013) higher in magnitude as compared to late variant epitope (LV) responses. There was no difference in polyfunction, memory maturation or perforin expression between EV and LV responses. Responses towards the mutant epitopes (ME) were observed before detectable sequence mutation (0.15-6% of total memory CD8 cells), but were mostly of lower magnitude and polyfunction as compared to EV responses. However, the profile of ME responses were highly variable amongst studied subjects and some mutant epitopes were recognized as efficiently (magnitude and polyfunction) as the transmitted sequences. After epitope mutation, responses towards EV epitopes decrease and no re-emergence of T cell reactivity to the ME was observed. Conclusion: Our data suggest that the magnitude of epitopespecific CD8+ T cells has more impact on selecting for epitope escape than multifunctionality or perforin expression. These data highlight that the phenotypic and functional profile of epitopespecific CD8+ T cells that imparts selective pressure on epitopes to mutate is likely to be a more complex interaction between CTL and the virus.

Background: A subset of T cells with immunosuppressive properties is the regulatory T cells (Treg), implicated in microbial infections. However, the role of Tregs in HIV-1 infection is controversial and two main hypotheses consider Tregs as 1) beneficial Tregs prevent chronic immune activation or 2) harmful Tregs suppress anti-HIV-1 immune responses. Also, T helper 17 (TH17) effector cells have been implicated in chronic inflammatory disease, but studies of TH17 cells in HIV-1 infection are few and conflicting. Furthermore, the balance between Tregs and TH17 cells appear to be important for disease outcome. To gain more information on the role of Tregs and TH17 cells in chronic HIV-1 infection, we evaluated their frequency and relationship in chronically infected HIV-1 patients with different control of infection. Methods: PBMC from 17 viremic individuals, 19 HAART patients, 13 Elite Controllers and 10 uninfected individuals were analyzed. Treg identification: PBMCs were subjected to staining and flow cytometry using antibodies against: CD3, CD4, CD25, CD127, FOXP3 and Live/Dead Fixable Dead Cell Stain. Identification of IL17A producing TH17 cells: PBMCs were stimulated with PMA/ionomycin in the presence of Brefeldin A and subjected to staining and flow cytometry using antibodies against: CD3, CD4, INFg, IL17A and Live/Dead Fixable Dead Cell Stain. Results: We demonstrate that Elite Controllers had lower levels of Tregs compared with HIV-1-infected viremic individuals but that the low Treg level did not differ between individuals with HIV-1 control, whether natural or therapy-induced. We also show that TH17/Treg ratio was similar in Elite Controllers and uninfected controls, whereas in viremic and treated HIV1-infected individuals the TH17/Treg ratio was lower compared with uninfected controls. Conclusion: We demonstrate that Elite Controllers have low level of Tregs compared with viremic individuals and that one characteristic of spontaneous HIV-1 control is a maintained balance between Tregs and TH17 cells.

AIDS Vaccine 2011

183

POSTERS

Posters
P17.21

Topic 17: T Cell Immunity P17.22


SIV Infection of Rhesus Macaques Results in Dysfunctional T and B Cell Responses to Neo and Recall Leishmania Major Vaccination
N.R. Klatt1, C.L. Vinton1, J. Ho2, P.A. Darrah3, J.D. Lifson4, S.L. Moir5, R.A. Seder6, J.M. Brenchley1
1

Influence of HAART on Alternative Reading Frame Immune Responses over the Course of HIV-1 Infection
S. Champiat1, N.J. Maness2, J. Lehman3, A. Hasenkrug1, J. Miller1, M. Hong4, J.N. Martin1, S.G. Deeks1, G.E. Spotts1, F.M. Hecht1, E.G. Kallas4, K.E. Garrison3, D.F. Nixon1
1

University of California, San Francisco, CA, USA; Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA; 3Saint Marys College of California, Moraga, CA, USA; 4University of So Paulo, So Paulo, Brazil
2

Background: Translational errors can result in bypassing of the main viral protein reading frames and the production of alternate reading frames (ARF). Within HIV-1, there are many such ARFs in both sense and the antisense directions of transcription. These ARFs have the potential to generate immunogenic peptides called cryptic epitopes (CE). Therapy and immune response exert a mutational pressure on HIV-1. Immune pressure exerted by ARF CD8+ T-cells on the virus has already been observed in vitro. HAART has also been described to select HIV-1 variants for drug escape mutations. Since the mutational pressure exerted on one location of the HIV-1 genome can potentially affect the 3 reading frames, we hypothesized that ARF responses would be affected by this drug pressure in vivo. Methods: In this study, we identified new CE derived from sense and antisense transcription of HIV-1 using an interferon-gamma elispot assay. To evaluate the influence of HAART on HIV-1, ARF T-cell responses over the course of HIV-1 infection were tested in acutely infected patients enrolled in a structured HAART interruption program and in chronically infected patients before and after HAART introduction. Results: We measured T cell responses to 199 HIV-1 CE encoded within 13 sense and 34 antisense HIV-1 alternative reading frames. We were able to observe that these ARF responses are more frequent and stronger in patients chronically infected compared to acutely infected, and that HAART increased the breadth of ARF responses. Conclusion: We confirmed that translation of ARF is an important source of T cell epitopes in HIV-1 infection. These CE responses are present both in the acute and the chronic phase of HIV-1 infection, but are stronger in chronically infected individuals. We observed that HAART modified the dynamics of ARF T-cell responses, suggesting that ARF T-cells may play a role in the emergence of virus mutation escape.

National Institutes of Health/National Institute of Allergy and Infectious Disease, Bethesda, MD, USA; 2National Institutes of Health/National Institute of Allergy anIAID/LIR, Bethesda, MD, USA; 3National Institutes of Health/National Institutes of Allergy andID/VRC, Bethesda, MD, USA; 4National Cancer Institute/ACVP/SAIC, Frederick, MD, USA; 5NIH/NIAID/LIR, Bethesda, MD, USA; 6National Institutes of Health/National Institute of Allergy and /VRC, Bethesda, MD, USA
Background: HIV infection is characterized by pathological immune activation and dysregulation of the immune system, including abnormal B and T-cell responses. While it is clear that lymphocytes are dysfunctional, and ultimately fail to protect HIV-infected individuals from opportunistic infections, the immunological mechanisms underlying lymphocytic dysfunctionality are unclear. Moreover, whether these immunological dysfunctions are restricted to memory or nave lymphocyte responses is unknown. Methods: Here we immunized SIV-infected and uninfected rhesus macaques to induce neo and recall responses to a Leishmania major polyprotein (MML) vaccine given with Poly(I:C) adjuvant. We measured the frequencies of antigen-specific T-cells by intracellular flow cytometric staining for TNF, IFNg, IL2, and CD40L and the frequencies of antigen-specific B-cells by IgA, IgM, and IgG ELISPOT. Results: We found that with the Poly(I:C) adjuvant, vaccinated SIVuninfected animals induced high frequencies of polyfunctional MML-specific CD4+ T-cells. However, in SIV infected animals, there was a significant loss of functionality in CD4+ T-cell responses to both neo MML vaccination (P=0.0011) and to recall MML vaccination (P=0.0082). Importantly, the dysfunctional MML-specific CD4+ T-cell response was not attributed to preferential SIV infection of these cells in vivo. Consistent with the loss of functionality we observed in CD4+ T-cells, we also found dysfunctional B-cell responses to MML vaccinations. After SIV infection, the frequency of MML-specific memory B-cells was decreased, with the greatest decrease observed in CD27+ memory B-cells that produced IgA in response to either neo (P=0.0221) or recall (P=0.0356) MML vaccinations. Conclusion: These data indicate that SIV infection results in dysfunctional T-cell responses to both neo and recall antigens and that dysfunctional CD4+ T-cell responses likely contribute to deficient B-cell responses. Furthermore, the robust responses observed using a Poly(I:C) adjuvant provides support for use of Poly(I:C) as an adjuvant for future vaccine design where high frequencies of antigen-specific T and B-cells are desired.

POSTERS 184 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.23


In Vitro Derived Dendritic Cells from CD34+ Precursors Share Features of Dendritic Cells from Several Tissues that Favour T Cell Immunity
G. Romain2, E. van Gulck1, O. Epaulard2, L. Adam2, N. Bosquet2, A. Cosma2, G. Vanham1, R. Le Grand2, F. Martinon2
1

Posters
P17.24
Delayed-Type Hypersensitivity (DTH) Elicited by Defined Cytotoxic T Lymphocyte (CTL) Epitopes as a Potential Immune Read-out for Vaccine Trials
M. Ruiz-Riol1, B. Mothe1, W. Howard2, B. Walker3, D. Scadden4, V. Snchez5, C. Brander 6 IrsiCaixa AIDS Research Institute HIVACAT, Barcelona, Spain; 2Hematologic Malignancies MGH Cancer Center, Massachusetts, MA, USA; 3Ragon Institute of MGH, MIT and Harvard, Massachusetts, MA, USA; 4Center for Regenerative Medicine, MGH, Boston, Massachusetts, MA, USA; 5IDIBAPSAIDS Research Group-HIVACAT, Hospital Clinic, Barcelona, Spain; 6Instituci Catalana de Recerca i Estudis Avancats, Barcelona, Spain
1

Retrovirology Research Unit, ITMA, Antwerp, Belgium; 2CEA / Universit Paris Sud, Fontenay aux Roses, France

Background: Dendritic cells (DCs) are heterogeneous cells spreading the body with a unique ability to prime nave T cells against invading antigens. In vitro derived DCs can be therefore considered for vaccination against HIV. Here, we assessed the resemblance between in vitro derived DCs and antigen-presenting cells (APCs) from several tissues, and we demonstrated its ability to induce anti-Gag T cell responses in cynomolgus macaques. Methods: Phenotype of DC subsets from cynomolgus macaque bone marrow (BM), blood, spleen, lymph nodes (LN) and epidermis as well as DCs derived in vitro from BM CD34+ progenitors (CD34-DCs) were studied by flow cytometry. CD34DCs were transfected with mRNA encoding HIV-Gag and injected into macaques by subcutaneous and intradermal routes. Immune response was studied by using ex vivo stimulated PBMC with pools of Gag peptides and by analyzing cytokine secretion in culture supernatants, the frequency of IFN- and IL-2 producing cells in ELISpots and the polyfunctional T cells by intra-cellular cytokine staining. Results: Three main APCs subsets reside in all tissues with the exception of the epidermis, which included only cells with Langerhans phenotype. CD34-DC cultures produced immature CD34-DCs sharing similarities with CD14+ APCs, and mature CD34DCs resembling CD11c+ myeloid DCs. These CD34-DCs efficiently stimulate T cells in vitro and vaccination of macaques with Gagexpressing CD34-DCs induced Gag specific CD4+ and CD8+ T cells producing IFN-, TNF-, MIP-1 and IL-2. Interestingly, the number of boost injections increased the frequency of polyfunctional CD8+ T cells and memory CD45RA- T cells. Conclusion: In vitro derived CD34-DCs shared features from both CD11c+ and CD14+ APC subsets. These characteristics might favour their high efficiency at inducing T cell immunity when used as vaccine vector.

Background: Assays that measure physiologically relevant effector function(s) of vaccine induced immune responses are a pre-requisite for effective HIV vaccine development. Immune responses to some vaccines are commonly tested by measuring cutaneous reactions to intradermal antigen injections. In most cases, entire protein antigens are used for this purpose. However, even short peptides can induce intra-dermal infiltration of CD4+ and CD8+ T-cells, suggesting that optimally-defined CTL cell epitopes could possibly elicit such delayed type hypersensitivity (DTH) responses on their own. Methods: In order to assess the potential to induce DTH reactions, 2 HLA-A*0201 restricted epitopes derived from HIV (SLYNTVATL, SL9) and Influenza (GILGFVFTL, GL9), respectively were used in 10 HIV infected (8 HLA-A*0201+) and 10 HIV uninfected (7 HLA-A*0201+) individuals. Local immune response (erythma and induration) were determined after 72h. Blood samples were obtained before injection and after 72h Results: No reactions were observed to SL9 injection while 6 of the 7 HIV negative and 5 of the 8 HIV positive HLA-A*0201 expressing individuals showed reaction to GL9 injection (induration diameter 3-15mm). HLA-A*0201 negative individuals did not show any reactivity. Among the HIV infected group, GL9 reactivity was not associated with CD4 counts or viral loads. Phenotypic analyses of epitope specific populations, including homing markers (CD103, CLA, CCR7, CXCR1 and LTBR) are currently being conducted to identify markers that could explain the differential migration potential of these epitope-specific CTL populations. Conclusion: Our data demonstrates that DTH reactions can be elicited by short CTL epitopes alone, in a HLA-dependent manner. The inability of HIV-specific cells induced upon natural infection to elicit a DTH reaction may reflect their impaired functionality in vivo. These data could help establish CTL-DTH as a simple, cheap and sensitive immune monitoring tool for large-scale vaccine trials for HIV and other pathogens.

AIDS Vaccine 2011

185

POSTERS

Posters
P17.25

Topic 17: T Cell Immunity P17.26


Enhancement of Gag-Specific but Reduction of Env- and Pol-Specific CD8+ T Cell Responses by SIV Regulatory and Accessory Proteins in Mice
S. Caijun1, Z. Yinfeng1, Z. Linqi2, C. Ling1
1

Conserved HIV-1 p24 Epitopes Elicit Cellular Immune Responses That Impact Disease Outcome
L.F. Tarosso1, V.A. Vieira1, E.G. Kallas1
1

School of Medicine, University of So Paulo, So Paulo, Brazil

Background: Breadth of HIV-1-specific cellular immune response and their impact on the control of viral replication have already been addressed by a number of studies. However, reported data have proven controversial. Here, we hypothesized that nature of targeted epitopes, rather than simply the total breadth or magnitude of HIV-1-specific responses, could clearer associate with disease outcome in infected patients. Methods: To address this issue, we investigated the possible presence of patterns of Gag p24 recognition among 27 HIV-1infected patients. We then attempted to explore epitope features that could define such patterns and how these patterns distinctly associate with disease progression. By ELISPOT-IFN-g assays, we screened mononuclear cells from HIV-1-infected subjects against Gag p24 15mer peptides encompassing the whole protein. Results: Obtained data was used to carry out a clustering analysis that unveiled the presence of two groups of patients with distinct patterns of Gag p24 recognition. Of note, despite targeted Gag p24 peptides were completely different between two groups, breadth and magnitude of the responses were not significantly different. Interestingly, viral control, measured as RNA copies in plasma, and preservation of CD4+ T cells were increased in one of the two groups. Additionally, we compared genetic conservation in the amino acid sequences of the peptides recognized by our patients, as well as the HLA-I-restricted epitopes within them. Markedly, subjects presenting higher control of HIV-1 replication were the ones targeting the more conserved epitopes. Furthermore, such higher genetic variation was present mainly in anchor residues for HLA-I molecules. Conclusion: Despite being already a concept among HIV vaccinologists, we show here, for the first time, experimental evidence from cases of the AIDS virus infection in humans that cellular immune responses targeting conserved HLA-I-restricted epitopes do associate to a better control of viral replication and maintenance of CD4+ T cells.

Guangzhou Institute and Biomedicine and Health (GIBH), Guangzhou, China; 2Comprehensive AIDS Research Center, Tsinghua University, Beijing, China
Background: Accessory and regulatory proteins (nonstructural proteins) have received increasing attention as components in novel HIV/SIV vaccine design. However, the complicated interactions between nonstructural proteins (Nef, Vif, Vpr, Vpx, Tat and Rev) and structural proteins(Gag, Pol and Env) remain poorly understood, especially their effects on immunogenicity. Methods: In this study, the immunogenicity of structural proteins with and without nonstructural proteins was compared. First, a series of recombinant plasmids and adenoviral vectors carrying various SIVmac239 nonstructural and structural genes were constructed. Then mice were primed with DNA plasmids and boosted with corresponding Ad5 vectors, and the resulting immune responses were measured. Results: Our results demonstrated that when the individual Gag, Pol or Env gene products were co-immunized with the whole repertoire of nonstructural proteins, the Gag-specific CD8+ T response was enhanced, while the Env and Pol-specific CD8+ T responses were significantly reduced. The same pattern was not observed in CD4+ T cell responses. Antibody responses against both the Gag and Env proteins were elicited more effectively when these structural antigens were immunized together with nonstructural antigens. Conclusion: These findings may provide useful guidance for designing novel HIV/SIV vaccine regimens that include nonstructural proteins as a component.

POSTERS 186 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.27


HIV Infection Induces the Alteration of Cytolytic Effector Molecule Expression Diversity in CD8+ T Cell Population
N. Onlamoon1, K. Sukapirom1, K. Polsrira1, K. Pattanapanyasat1
1 Faculty of Medicine Siriraj Hospital / Mahidol University, Bangkok, Thailand

Posters
P17.28 LB
Occult Replication of a Conditionally-Live Attenuated SIV Profoundly Upregulates T Effector Memory Cell Frequency
M. Manoussaka3, R. Stebbings1, N. Berry1, A. Das2, B. Berkhout2, N. Almond1, M.P. Cranage3 National Institute for Biological Standards and Control, Potters Bar, United Kingdom (Great Britain); 2Academic Medical Center of the University of Amsterdam, Amsterdam, Netherlands; 3St Georges University of London, London, United Kingdom (Great Britain)
1

Background: Although many studies reported that cytolytic function of HIV-specific CD8+ T cells are often impaired during the course of HIV infection, it remains unclear whether this functional failure is selective for HIV-specific population or an overall phenomenon. An analysis of a bulk CD8+ T cell population can improve our understanding of a neglected area of CD8+ T cell impairment that stretches far beyond HIV-specific CD8+ T cell population. Methods: In this study, we determined the diversity of CD8+ T cells in term of cytolytic molecule expression including perforin (Pr), granzyme A (GrA) and B (GrB) by using intracellular staining and flow cytometry technique. The results were compared between healthy individuals and untreated HIV infected patients. Correlations with markers of disease progression including the percentage of CD4+ T cells, absolute CD4 count and viral load were also determined in this study. Results: We demonstrated the presence of three different subsets of CD8+ T cells that expressed different combinations of cytolytic effector molecules including triple (Pr+GrA+GrB+), double (PrGrA+GrB+) and single (Pr-GrA+GrB-) positive subsets. Results showed significant increase in the frequencies of triple and double positive subsets whereas a decrease in single positive subset was observed in HIV infected patients when compared to healthy individuals. Furthermore, a positive correlation between the frequency of triple positive subset and viral load was observed whereas a negative correlation between the frequency of single positive subset and viral load was identified. Conclusion: We demonstrated that the diversity of CD8+ T cell subsets based on co-expression of cytolytic effector molecules changed during HIV infection and this alteration correlated with stages of disease progression. However, the mechanism that initiates this subset alteration remains unclear.

Background: The most potent protection against infection with virulent SIV, including protection against mucosal challenge, is conferred by vaccination with live attenuated virus. Although this approach is precluded for HIV because of safety concerns, understanding the mechanisms of superinfection resistance may inform rational vaccine design. Methods: In order to uncouple antigen exposure from active viral replication we compared peripheral and intestinal T cell phenotype and SIV peptide-specific responses following infection of macaques with wild type SIVmac239, attenuated SIVmac239nef or with a doxycycline (dox)-conditional replication variant of SIVmac239 nef designated SIVrtTA. Global (antigen-nonspecific) T cell phenotype was assessed for central memory (Tcm) (CD28+ CD95+) and effector memory (Tem) (CD28- CD95+) and SIV-specific T cell responses were measured by detection of TNF-, IFN-, IL-2 and IL-17. Results: In animals in which on-going virus replication was permitted (SIVrtTA + dox & SIVmac239nef), the proportion of CD4 and CD8 Tcm were reduced in the majority of animals while Tem proportions increased. In animals infected with SIVrtTA in which dox had been withdrawn for 8 weeks prior to analysis, these changes were not seen. Moreover animals infected with wild type virus had elevated CD4 and CD8 Tcm. Analysis of gut mucosal homing (4+7+ and 7+) T cells showed similar polarised changes. Conclusion: Overall, we found that active replication of SIVrtTA and SIVmac239nef had a profound impact on global T cell phenotype and antigen-specific polyfunctionality in the periphery and the gut. The use of these SIV mutants will contribute to the understanding of the mechanisms of superinfection resistance

AIDS Vaccine 2011

187

POSTERS

Posters
P17.29 LB

Topic 17: T Cell Immunity P17.30 LB


Senegal Multi-Site Evaluation of the PointOf-Care PIMA Instrument for CD4+ T-Cell Enumeration Using Venous and Capillary Blood
P. Diaw1, G. Daneau2, I.P. Ndiaye1, A.A. Coly1, M. Camara1, S. Mboup1, L. Kestens2 LBV-RARS Senegal, Dakar, Senegal; 2Institut De Medecine Tropical, Anvers, Belgium
1

In Vivo Electroporation Induces Broad HIV-1 Envelope Epitope Responses to ADVAX HIV-1 DNA Vaccine in Humans
J. Kopycinski1, H. Cheeseman1, A. Ashraf1, D.K. Gill1, P. Hayes1, M. De Souza2, P. Fast1, J.H. Cox1, D. Hannaman3, J. Gilmour1, S. Vasan4
1

International AIDS Vaccine Initiative, London, United Kingdom (Great Britain); 2Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 3Ichor Medical Systems, Inc, San Diego, CA, USA; 4Aaron Diamond AIDS Research Center, New York, NY, USA
Background: DNA-based vaccines have been found safe, stable, and are unencumbered by pre-existing vector-specific immune responses, although previous clinical studies suggested they were poorly immunogenic. Administration of the HIV1 clade B/C ADVAX DNA vaccine (Gag-Pol, Nef-Tat and Env) by electroporation has been shown to be safe and to result in increased and more durable cellular immunogenicity over intramuscular administration. The majority of responses induced were of a polyfunctional phenotype in CD4+ T-cell populations. Here, we investigate the breadth of T-cell responses against the ADVAX envelope (gp160) protein along with the identities of regions recognized through epitope mapping. Methods: PBMCs isolated from 16 recipients of ADVAX EP were selected based on ELISpot responses >100 SFU/106 PBMC and subjected to peptide mapping using a matrix containing sequences corresponding to the ADVAX gp160 insert. Potential positive peptides were deconvoluted and, whenever possible, responses confirmed using short-term T-cell lines generated following 9-day culture with the whole gp160 pool with IL-2. Results: Following deconvolution in 11/16 individuals, the breadth of gp160-specific responses was comparable to the epitope breadth induced by MVA and Ad5 vectors in other trials (mean breadth = 2.8 epitopes/volunteer). The majority of responses focused on V3/C4 and V2 regions. 6/11 volunteers tested recognized the a4b7 integrin-binding domain of gp120. A trend towards an association with increased vaccine dose and breadth was seen. Conclusion: Together these data indicate that electroporation of the ADVAX DNA vaccine induces T-cell responses towards multiple regions of gp160, including the alpha 4 beta 7 integrin binding site. Cellular and humoral immunogenicity may be further enhanced through combination with adjuvants and/or boosting with viral vector or protein vaccines.

Background: CD4+ T-cell enumeration is used as a criterion to initiate antiretroviral therapy (ART) in HIV patients and to monitor treatment efficacy. However, simple, affordable and reliable point-of-care instruments adapted to resource-limited settings are still lacking. PIMA is a new point-of-care CD4 instrument using disposable cartridges and battery-powered analyzer. Methods: Whole blood samples were taken by venipuncture or by fingerprick, from 300 subjects, including HIV-infected patients and HIV(-) controls. CD4+ T-cell counts were measured on PIMA (using venous or capillary blood) and on FACSCount (using venous blood, considered as the reference). External quality controls were from AFREQUAS. Results: CD4+ T-cell counts were similar on PIMA and FACSCount using either venous blood or fingerprick samples (concordance correlation coefficient 0.93 and 0.88 respectively), with better accuracy for low CD4+ T-cell counts. Considering clinical decision to start ART at 200 CD4+ T-cells/l, kappa coefficient for agreement was 87% for venous and 85% for capillary blood, and 78 and 71% respectively at a 350 CD4+ T-cells/l cut-off. Sensitivity was 90%/91% and specificity 98%/96% at 200cells/ l for venous/capillary blood respectively, and 98%/91% and 79%/80% at 350cells/l. Repeatability (precision) on venous blood resulted in a coefficient of variation (CV) of 4%. Using fingerprick blood, the error frequency (aborted analysis) was 14%. Conclusion: PIMA device is of good value to follow-up adult HIV-infected patients in resource-limited settings. Besides affordability, easy and safe practice, we demonstrated that performance was similar to FACSCount for relevant clinical CD4+ T-cell counts. However, technical error prevention requires good training for fingerprick samples.

POSTERS 188 AIDS Vaccine 2011

Topic 17: T Cell Immunity P17.31 LB


Efficacious SIV-Specific CD8+ T Cell Responses in Heterozygous Animals are Weighted Toward Peptides Presented by a Single MHC Haplotype
J.M. Greene1, E.N. Chin1, J.J. Lhost1, M.L. Budde1, P.J. Hines1, B.J. Burwitz1, D.H. OConnor1
1

Posters
P17.32 LB
Primary Immune Responses to Vaccinia Virus Vaccination: The Role of Cytotoxic Effector CD4+ T Cells in the Generation of Human T Cell Memory
C.M. Munier1, S. Ip1, M. Bailey1, Y. Xu1, S. Liu2, D.A. Cooper1, S.J. Kent3, J.J. Zaunders4, A.D. Kelleher1
1

University of Wisconsin - Madison, Madison, WI, USA

Background: Certain MHC class I alleles are correlated with remarkable control of HIV and SIV, indicating that specific CD8+ T cell responses can effectively control viral replication; however, we do not understand why some individuals with these MHC class I alleles do not control their viral loads. Methods: We used animals with defined MHC haplotypes (A and B) and viral inhibition assays to directly assess CD8+ T cell efficacy ex vivo. We isolated CD8+ T cells and CD8-depleted target cells from the blood of six homozygous AA, six homozygous BB and six heterozygous AB animals. We added CD8+ T cells to infected target cells in every possible combination. We then measured the ability for CD8+ T cells to reduce viral replication through intracellular p27 staining. Results: While CD8+ T cells from homozygous AA, BB, and heterozygous AB animals were equally capable of suppressing viral replication on autologous and AB target cells, we found that AB heterozygotes suppress viral replication most effectively on AA target cells. Surprisingly, the same AB effector cells did not effectively inhibit viral replication on BB target cells, revealing a striking absence of effective B restricted CD8+ T cell responses. Conclusion: These results indicate that the greater potential breadth of CD8+ T cell responses present in heterozygous animals does not necessarily lead to greater antiviral efficacy and suggest that CD8+ T cell responses in heterozygous animals have a skewed focus toward epitopes restricted by a single haplotype. Such MHC haplodominance may help explain why only some individuals with protective MHC class I alleles durably control HIV replication.

The Kirby Institute and St Vincents Centre for Applied Medical Research, Sydney, Australia; 2The Garvan Institute, Sydney, Australia; 3The University of Melbourne, Melbourne, Australia; 4St Vincents Centre for Applied Medical Research, Sydney, Australia

Background: To understand the developmental stages of CD4+ T cell responses to viral infection and their differentiation into long-term memory cells, we studied individuals receiving primary vaccinia virus (VV) vaccination. This provides an attractive model for the study of human antiviral T cell responses as vaccination results in an acute infection that is cleared and leads to long-term protective immunity. Methods: At the peak of the primary effector cell response to VV, day 13 post-vaccination, we purified activated effector (CD45RO+CD38+++) and nave (CD45RO-CD38dim) CD4+ T cells from 2 subjects, extracted mRNA and conducted microarray analysis using the Affymetrix, HU133 Plus 2.0 microarray. Results: In the activated effector CD4+ compared to nave CD4+ T cells many of the genes up-regulated were associated with cell division and activation, such as Ki-67, CD38 and CCR5, as expected, hence validating the arrays. Surprisingly, there was a strong up-regulation of cytotoxic T-lymphocyte (CTL) associated genes in the activated effector CD4+ T cells. Of the top 40 differentially expressed genes granzyme K ranked 1st with fold change (fc) of 40 compared to nave CD4+ T cells. Killer cell lectin-like receptor subfamily B member 1 (KLRB1/CD161):4th (fc:26.3), Rab27a:19th (fc:8.2), granzyme A:26th (fc:13.9) and granulysin:36th (fc:7.9). These fold changes are being validated by qPCR. Preliminary analysis at 13 days post vaccination with VV revealed >10% of activated VV-specific CD4+ T cells and >50% of activated CD8+ T cells expressed granzyme K. Conclusion: Generation of anti-viral human CD4+ T cell memory during primary immune responses is poorly understood. The role of CTL associated genes in this process has not been explored. Understanding their role in the generation of an effective memory may lead us closer to the development of a more effective HIV vaccine, such as enhancing the modestly efficacious HIV-1 vaccine used in the RV144 study.

AIDS Vaccine 2011

189

POSTERS

Posters
P17.33 LB

Topic 17: T Cell Immunity

Ubiquitination of the HIV Gag Protein Results in an Increased Expression of Inhibitory Markers on Stimulated T Cells
S. Herath1, A. Benlahrech1, T. Athanasopoulos2, C. Hervouet3, P. Becker3, L. Klavinskis3, G. Dickson2, A. Meiser1, S. Patterson1 Imperial College, London, United Kingdom (Great Britain); Royal Holloway, Egham, United Kingdom (Great Britain); 3 Kings College, London, United Kingdom (Great Britain)
1 2

Background: The development of an HIV vaccine that generates a large pool of CD8+ T cells recognizing multiple cytotoxic lymphocyte (CTL) epitopes is a primary strategy in HIV vaccination protocols. Broadening of the CTL repertoire ensures responses to HIV proteins that are able to rapidly mutate as a result of selective pressures imposed by the immune system. Consequently, our strategy employed ubiquitination of the gag protein, with the aim of targeting the protein to the proteasome, thus enhancing MHC class I antigen processing and thereby broadening the CD8 T cell repertoire. Methods: T cells were primed and boosted weekly with monocyte derived dendritic cells (mDCs) transfected with either rAd5 CN54 HIV ubiquitinated gag or rAd5 CN54 HIV non-ubiquitinated gag for a total of 4 weeks, and then assayed for antigen-specific responses by restimulation with peptides. The production of IFN gamma was determined by ELISPOT and intracellular staining and differences in activation status were determined by the expression of surface markers by flow Results: Although ubiquitination enhanced proteasomal targeting, unexpectedly, IFN gamma production was diminished in T cells primed and boosted against ubiquitinated gag proteins. To understand the absence of an enhanced T cell response, we investigated the expression of several markers, including CD38, TIM3, and CTLA-4 by flow cytometry. Preliminary results indicated that at 4 weeks post in vitro culture, T cells primed against ubiquitinated gag proteins and restimulated with peptides, expressed an increased frequency of some inhibitory markers as compared to T cells primed against non-ubiquitinated gag proteins. Interestingly, expression of inhibitory markers did not influence the proliferative capacity of lymphocytes as cell numbers increased progressively. Conclusion: These results suggest that ubiquitination of the HIV gag protein may lead to chronic stimulation of T cells in vitro, which may lead to their exhaustion and consequently their reduced responsive capacity.

POSTERS 190 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.01


Optimizing Delivery of the HIVconsv Immunogen in Rhesus Macaques
M. Rosario1, A. Mbewe2, E.D. Quakkelaar3, C.J. Melief3, T. Hanke4 Li Ka Shing Institute of Virology University of Alberta, Edmonton, Canada; 2Weatherall Institute of Molecular Medicine, Oxford, United Kingdom (Great Britain); 3Leiden University Medical Centre, Leiden, Netherlands; 4The Jenner Institute, Oxford, United Kingdom (Great Britain)
1

Posters

P18.02
Immune Selection of T Helper Epitopes in Healthy Volunteers who Received a Multiepitope Candidate HIV Vaccine
H. Chen4, E. Kennedy1, M. Newman2, M. Keefer4, B. Kim3, A. Sant3, X. Jin4 Departments of Microbiology and Immunology, University of Rochester, Rochester, NY, USA; 2GeovVx Inc., Atlanta, GA, USA; 3 Departments of Microbiology and Immunology, University of Rochester, USA; 4University of Rochester, Rochester, NY, USA
1

Background: A safe and effective vaccine remains the best hope for halting the HIV-1 pandemic. Here we follow-on from a previous macaque trial to further define the responses in which HIVconsv, an immunogen which encodes the most conserved regions of clades A-D, was administered as SLP and shown to increase the breath of T-cell responses to HIVconsv in three Mamu-A*01+ rhesus macaques. Methods: Three groups of five rhesus macaques were vaccinated with HIVconsv. HIVconsv was vectored in plasmid DNA for electroporation(D), chimpanzee adenovirus(C), modified vaccinia virus Ankara(M), and in synthetic long peptides(S). Each group received either SSSCMS, DSSCMS, or DDDCMS regimen and were followed for qualitative and quantitative T-cell responses. The DDDCMS group was further vaccinated with HIVconsv vectored in Bacillus Calmette-Gurin mycoplasma(B). Results: SLP vaccination did not induce robust responses against HIVconsv even after prior administration of plasmid DNA however, electroporation did spare the amount of DNA needed. The DDDCM regimen mimicked a DDDAM(A= Human adenovirus HAdV5) regimen previously shown to be highly immunogenic in macaques and also being trialled in humans. Next, the CD4 T-cell compartment was determined to be the major responder to SLP.HIVconsv vaccination increasing in T-cell breath and proliferative response. Multiple cytokine production from both central memory and effector memory T-cell subsets were also augmented by SLP vaccination. Finally, boosting with HIVconsv vectored in BCG further enhanced the polyfunctionality of both central and effector memory T-cell responses more so than the other vectors. Conclusion: Our macaque trial clearly demonstrates the potency of SLP to generate broad CD4 T-cell responses with a polyfunctional profile against conserved regions of HIV-1 in rhesus macaques when administered in a strict regimen. Because SLP vaccination, in this regimen, greatly expands the CD4 T-cell compartment responding to HIVconsv, it facilitates the possibility of amalgamating an antibody vaccine into an effective CTL vaccine.

Background: Previously, we detected helper T lymphocyte (HTL) responses in 27/39 individuals two weeks after the 4th vaccination with a recombinant protein HIV vaccine comprised of 18 HTL epitopes in a Phase I clinical trial, HVTN064. In current study, we examined the individual epitope-specific responses and correlated these responses with predicted protein structure. Methods: PBMC from the 27 responders were stimulated for 2022 hours by individual peptide epitopes, and IFN, TNF, IL-2, IL4, IL-5 and IL-10 were measured in culture supernatants with the Cytometric Bead Array (CBA) assay. We used PSIPRED to predict secondary structure for the entire HTL vaccine polypeptide, and examined the association between protein secondary structure and epitope selection. Results: All 18 epitopes were recognized (>mean+3SD of controls) in at least one of the 27 individuals, 14 epitopes were recognized in more than 30% of individuals, and 5 epitopes were recognized in more than 50% of individuals. Of the 9 epitopes that stimulated IL-2, IFN, and IL-5 responses in vaccinated mice, 8 stimulated secretion of these three cytokines in more than 20% of human vaccine recipients. Among 9 immunodominant epitopes that stimulated a triple cytokine response in more than 30% of subjects, 6/9 sit in the regions of strand and/or coil, while 3/9 were in the helix regions. Additionally, 3 of the epitopes were positioned on either ends of the protein vaccine. Conclusion: We found that half (9/18) of the predicted HIV HTL epitopes elicited strong immunity when tested as part of a vaccine, and protein structure analysis did not completely predict the immune response hierarchy. Thus, current methods of HIV HTL multiepitope vaccine design and preclinical testing in mice, while valuable, do not translate entirely into human studies, therefore improved methods for HTL vaccine design and testing are needed.

AIDS Vaccine 2011

191

POSTERS

Posters
P18.03

Topic 18: T Cell Vaccine Concepts and Design P18.04


Low Dose Immunization of Codon-Optimized Recombinant BCG Confers Long-Lasting SIVSpecific Memory T Cells After Recombinant Vaccinia Boost in Macaques
M. Kanekiyo1, Y. Ami2, K. Matsuo3, K. Someya2, N. Yamamoto2, M. Honda2
1

DNA Boosting Elicited Broader T Cell Responses Against HIV-1 Tat-Rev-Integrase(C-half)-Vif-Nef Fusion Gene Vaccine than Recombinant Vaccinia Boosting
Y. Wan1
1

Shanghai Public Health Clinical Center, Shanghai, China

Background: We previously compared the immunogenicities of DNA vaccines containing tat-rev-integrase(C-half)-vif-nef fusion genes derived from the top three of the most prevalent HIV-1 clades in China, including B`, B`/C and AE recombinant subtype of HIV-1. And our data showed that Integrase(C-half) and Vif were consistently the most immunogenic components in three DNA vaccines. As both cellular immune breadth and depth are crucial for the efficacy of T cell based HIV-1 vaccine, we were thus curious about whether the T cell responses against Tat, Rev and Nef could be improved. Methods: Three recombinant Tiantan vaccinia vectored vaccines were constructed respectively by using polynecleotides encompassing tat, rev, integrase(C-half), vif and nef genes(TRIVN) of HIV-1cn54(B`/C),HIV-1rl42(B`) and HIV-1ae2f(AE), which had been designed and synthesized previously. Female BALB/c mice were immunized in either DNA priming-DNA boosting or DNA priming-rTTV boosting regimen. IFN- Elispot assay was used to read out the specific T cell immunity. Results: The regimen of mixed DNA priming-mixed DNA boosting seemed to be able to elicit comparable magnitude of specific T cell responses against HIV-1 consensus B peptides with regimen of mixed DNA priming-mixed rTTV boosting (500155 SFCs/106 splenocytes vs 430173 SFCs/106 splenocytes). Both were significantly higher than those induced by pSV-TRIVN(rl42) and pSV-TRIVN(cn54). And more interestingly, our data showed the mixed DNA-mixed DNA regimen could enhance T cell responses against sub-dominant compents of the fusion gene vaccine, e.g. peptides of consensus B Tat and HIV-1 AE Nef. While, when boosting with mixed rTTV, specific T cell responses tended to be more concentrated upon immune dominant constituents, including Integrase(C-half) and Vif. Conclusion: Our data suggested that boosted with mixed DNA vaccine could somehow elicit broader T cell responses against less immunogenic parts of tat-rev-integrase(C-half)-vif-nef fusion gene vaccine.

National Institute of Infectious Diseases (VRC, NIH), Tokyo, Japan; 2National Institute of Infectious Diseases, Tokyo, Japan; 3Japan BCG Laboratory, Tokyo, Japan

Background: Mycobacterium bovis bacillus Calmette-Gurin (BCG) has been extensively studied as a primer of heterologous vaccination regimens because of its safety record, affordability and easy antigen delivery to the professional antigen presenting cells, and thereby to the T cells. As we reported, a prime-boost regimen combining recombinant BCG (rBCG)-SIVgag with a nonreplicating vaccinia virus DIs (rDIs-SIVgag) induced effective cellular immunity that was able to control a highly pathogenic SHIV after mucosal challenge in macaques. Methods: rBCG harboring the codon-optimized SIV gag gene (rBCG-SIVgag-opt) with a 10-fold higher expression than the native gag gene (rBCG-SIVgag) was constructed. Cynomolgus macaques were immunized with a low dose (106 bacilli) of this construct or rBCG-SIVgag, respectively, and then boosted with 107 plaque-forming-unit of rDIs-SIVgag twice at 30 and 61 weeks post-priming. After 3 years of the second boost, the animals were challenged with high dose of SIVmac 239 intrarectally and plasma viral load and CD4+cell counts in PBMC were monitored. SIV Gag-specific cellular responses were evaluated by interferongamma ELISPOT and flow cytometric analyses. Results: After rDIs-boost, rBCG-SIVgag-opt-primed macaques exhibited higher CD4+ and CD8+ T cell responses than those in rBCG-SIVgag-primed ones and showed significant Gag-specific CD8+ T cell recall responses even after 3-years interval. Two out of 3 animals in the rBCG-SIVgag-opt-primed group showed about 2 log reduction of plasma viral load at set-point in comparison with those in the control group. Conclusion: A low dose of rBCG-SIVgag-opt induced optimal priming of Gag-specific T cells and prolonged the maintenance of memory T cell response after vaccinia DIs boost. These results imply that the quality of the priming vaccine is a critical factor for inducing a desirable immune response against immunodeficiency viruses.

POSTERS 192 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.05


Boosting of Replication Competent NYVAC Primed HIV-1-Specific T-Cell Responses by HIV Synthetic Long Peptides (SLP)
P. Mooij1, I. Nieuwenhuis1, N. Beenhakker1, E.D. Quakkelaar2, J.L. Heeney3, M. Esteban4, B. Jacobs5, G. Pantaleo6, W.M. Bogers1, C. Melief7, G. Koopman1 Biomedical Primate Research Centre, Rijswijk, Netherlands; 2 Leiden University Medical Center, Leiden, Netherlands; 3 Cambridge University, Cambridge, United Kingdom (Great Britain); 4Centro Nacional de Biotecnologa, Madrid, Spain; 5 Arizona State University, Tempe, AZ, USA; 6University of Lausanne, Lausanne, Switzerland; 7LUMC, Leiden, Netherlands
1

Posters

P18.06
Characterization of Cellular Immune Responses Elicited by Ad35-GRIN/ENV and MVA Construct Prime-Boost Regimens
J. Cox1, S. Ratto-Kim2, J.R. Currier2, J. Excler1, E. Sayeed1, J.H. Kim2, M. Marovich2
1 2

International AIDS Vaccine Initiative, Rockville, MD, USA; U.S. Military HIV Research Program, Rockville, MD, USA

Background: We characterized prime-boost vaccine regimens using heterologous and homologous vector and gene inserts. Heterologous regimens offer a promising approach to focusing the cell-mediated immune response on the insert and away from vector-dominated responses. Methods: Ad35-GRIN/ENV vaccine is comprised of two vectors containing sequences from HIV-1 subtype A (GRIN=gag, rt, int and nef and ENV=env). MVA-CMDR, MVA-KEA and MVA-TZC vaccines contain gag, env and pol genes from HIV-1 subtype CRF01_AE, subtype A and subtype C, respectively. Balb/c mice were immunized with different heterologous and homologous vector and insert prime-boost combinations. Mice were examined for HIV and vector-specific immune responses on days 3, 7, 14 and 28 post-boost. Gag-specific IFN-g ELISPOT, ICS (CD107a, IFN-g, TNFa and IL-2), pentamer staining, and T-cell memory markers were used to differentiate responses to homologous vs. heterologous inserts. Results: Ad35-GRIN/ENV prime followed by boost with any of the MVA constructs induced Gag-specific responses superior to MVA prime-Ad35 boost or homologous Ad35-Ad35 or MVA-MVA prime-boost combinations. Notably, there was a dramatic shift from a vector focused response in homologous vector prime-boost regimens to an insert focused response in the heterologous vector prime-boost regimens (0.78% CD8+IFN-g_ to Gag and 4.24% CD8+IFN-g_ to MVAp581 in MVA-CMDR/MVA-CMDR vs. 5.74% CD8+IFN-g to Gag and 0.61% CD8+IFN-g_ to MVAp581 in Ad35-GRIN/ENV-MVACMDR). Gag-specific central and effector memory T cells were detected earlier and in greater frequency in the heterologous prime-boost regimens. IFN-g ELISPOT and ICS responses to other vaccine proteins showed similar response patterns. Conclusion: These results suggest that heterologous primeboost vaccination regimens can direct and enhance immunity by increasing the magnitude and polyfunctionality of an insertspecific cell-mediated immune response that is more rapidly detectable, compared to homologous vaccination regimens. This study supports the rationale and current plans for testing heterologous prime-boost regimens in clinical trials.

Background: To enhance the induction of insert specific immune responses, a new generation of replication competent poxvirus vectors was designed and evaluated against non-replicating poxvirus vectors in a HIV vaccine study in non human primates. Methods: Rhesus macaques were immunized with either the non-replicating variant NYVAC-GagPolNef HIV-1 clade C or the replicating NYVAC-GagPolNef-C-KC, boosted with HIVGagPolEnv-SLP and immune responses were monitored. Results: Gag-specific T-cell responses were only detected in animals immunized with the replicating NYVAC-GagPolNef-CKC variant. Further enhancement and broadening of the immune response was studied by boosting the animals with novel T-cell immunogens HIVconsv synthetic long peptides (SLP), which direct vaccine-induced responses to the most conserved regions of HIV and contain both CD4 T-helper and CD8 CTL epitopes. The adjuvanted (Montanide ISA-720) SLP divided into subpools and delivered into anatomically separate sites enhanced the Gag-specific T-cell responses in 4 out of 6 animals, to more than 1000 SFC/106 PBMC in some animals. Furthermore, the SLP immunization broadened the immune response in 4 out of 6 animals to multiple Pol epitopes. Even Env-specific responses, to which the animals had not been primed, were induced by SLP in 2 out of 6 animals. Conclusion: This new immunization strategy of priming with replicating competent poxvirus NYVAC-HIVGagPolNef and boosting with HIVGagPolEnv-SLP, induced strong and broad T-cell responses and provides a promising new HIV vaccine approach. This study was performed within the Poxvirus T-cell Vaccine Discovery Consortium (PTVDC) which is part of the CAVD program.

AIDS Vaccine 2011

193

POSTERS

Posters
P18.07

Topic 18: T Cell Vaccine Concepts and Design P18.08


IL-12 and GM-CSF Administration During the Prime in DNA/MVA Schemes Improves Magnitude, Breadth and Avidity of Cellular Response Against NefBF
A. Rodriguez1, C. Maeto1, M. Pascutti1, J. Falivene1, G. Turk1, M. Gherardi1
1

Optimization of the MVA Vaccine Potential After Deletion of a Viral Gene Coding for the IL-18 Binding Protein
J. Falivene1, M.P. Del Medico Zajac2, A.M. Rodrguez1, M.F. Pascutti1, C.A. Maeto1, B. Perdiguero3, C. Gmez3, M. Esteban3, G. Calamante2, M.M. Gherardi1
1

Centro Nacional de Referencia para el SIDA, Buenos Aires, Argentina; 2Instituto de Biotecnologa, CICVyA, INTA Castelar, Buenos Aires, Argentina; 3Centro Nacional de Biotecnologa, Depart. Biologa Molecular y Celular, Madrid, Spain

Centro Nacional de Referencia para el SIDA, Buenos Aires, Argentina

Background: Modified Vaccinia Ankara (MVA) is an attenuated strain of Vaccinia virus (VV) currently employed in many clinical trials against HIV/AIDS. MVA still retains genes involved in host immune response evasion, enabling its optimization by removing some of them. The aim of this work was to evaluate cellular immune responses (CIR) induced by a MVA bearing an IL-18 binding protein gene deletion (MVAIL-18bp). Methods: Balb/c and C57Bl/6 mice were immunized with different doses of MVAIL-18bp or MVA wild type (MVAwt), then CIR to VV epitopes were evaluated in spleen and draining lymph nodes at acute and memory phases (7 and 40 days postimmunization respectively). The proportion of IFN-gamma and IL-2 producing cells were measured by ELISPOT. The percentage of cytotoxic TCD8+ cells was analyzed by flow cytometry through CD107a/b surface marker. In vivo protection against an intranasal challenge with the replicative Western Reserve (WR) strain was evaluated during the memory phase, measuring the weight of individual Balb/c mice during two weeks. Finally, CIR against HIV-1 antigens expressed from MVA vectors was analyzed by ELISPOT using DNA prime/MVA boost schemes. Results: Compared with MVAwt, MVAIL-18bp immunization induced a significant two or three-fold increase in TCD8+ (p<0.01) and TCD4+ (p<0.05) responses to different VV epitopes, and increased the percentage of cytotoxic TCD8+ cells (p<0.05) in the acute phase. Potentiation of MVAs immunogenicity was also observed in the memory phase, in correlation with a higher protection against an intranasal challenge with WR. More importantly we also observed a significant increase in the CIR against HIV antigens such as Env, Gag, Pol and Nef from different subtypes expressed from MVAIL-18bp in DNA prime/ MVA boost vaccination regimens. Conclusion: IL-18bp contributes to immune response evasion during MVA infection, as its deletion from the viral genome potentiated the MVA immunogenicity observed against vector antigens and more importantly to HIV antigens.

Background: In Argentina, the HIV epidemic is characterized by the co-circulation of subtype B and BF variants, mostly CRF12_ BF. Nef is a highly variable protein among subtypes, making it a good tool to study the impact of HIV-1 inter-variant variability in the vaccine design setting (a difference between B and BF:24%). We previously reported a highly specific response against NefBF with low cross-reactivity to NefB in mice. The aim of this work was to analyze the possibility to improve the immune response induced by the co-delivery of cytokines during the priming doses. Methods: Mice received an intramuscular prime: 3xDNAnefBF (Group I, GI), 3xDNAnefBF+DNA-IL-12 (GII), or 3xDNAnefBF+DNA-GM-CSF (GIII), or 3xDNAnefBF+DNA-IL12+DNA-GM-CSF (GIV). Afterwards, all the groups received MVAnefBF as intraperitoneal boost. Nine days after the last immunization, the specific cellular immune response (CIR) was evaluated in the spleen using overlapping peptides representing NefBF or NefB by ELISPOT. Results: The highest responses were detected in GII (p=0.0317) and GIV. The observed increments vs. GI were (median [range]): GII 2.38 [1.21-2.71], GIV 1.535 [1.05-2.86]. After fine mapping the response, the peptides targeted were identified: all groups recognized two peptides, located on the N-terminal (BF) and the loop of the protein (BF and crossreactivity against B). GII and GIV also recognized a peptide located on the central core region (BF). Even more, GII recognized the B peptide with a frequency higher than the other groups, showing an enhanced cross-reactivity. We evaluated the cell avidity against the loop peptide (comparing sequences BF vs. B). We found that GII showed a higher avidity against the peptide B compared to GI (p=0.0011), whereas in both groups the highest avidity was detected against the peptide BF (BF vs. B: p<0.001). Conclusion: The use of cytokines in DNA/MVA schemes improved the CIR, incrementing the magnitude, avidity and breadth (B cross-reactivity) of the response induced.

POSTERS 194 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.09


Deletion of Genes B8R and A52L Enhances Vaccinia Virus Tiantan Immunogenicity and Attenuates Neurovirulence in Mice
Y. Liu1, D. Ling1, M. Liu1, Y. Ma1, Z. Liu1, M. Tian1, H. Peng1, D. Duan1, Y. Shao1
1

Posters

P18.10
A Novel HIV-1 Vaccine Strategy Using Live Recombinant Vesicular Stomatitis Viruses Expressing HIV-1 Protease Cleavage Sites
D. Tang1, R. Capina1, C. Semeniuk1, X. Yuan1, A. Krawchenko1, G. Kobinger1, F. Plummer1, M. Luo1
1

National Center for AIDS/STD Prevention and Control, Chinese Center, Beijing, China

Public Health Agency of Canada, Winnipeg, Canada

Background: Vaccinia virus Tiantan (vTT) is the vaccine that used to eradicate smallpox in China and is being developed as a recombinant vaccine for AIDS. Removal of specific immunomodulatory genes encoded by Tiantan may improve its potential as a vaccine. Protein A52 ( referred as WR A41) has sequence similarity to the VACV chemokine-binding protein and associated with reduced inflammation during dermal infection. B8R gene encodes a secreted protein with homology to IFN-gamma receptor, which neutralizes the antiviral and immunological regulation activities of IFN-gamma. Methods: The deletion mutants vTTB8R/A52L was engineered from VTT containing B8R and A52L gene deletions. HIV genes (gag, pol and gp145) from HIV-1 B/C 97CN54 were inserted into the TK region of vTTB8R/A52L. To assess the pathogenicity of the deletion mutant, groups of female BALB/c mice were infected with vTTB8R/A52L or vTT by the intranasal and intracephalic routes. A DNA prime/vaccinia boost regime was used to compare adaptive and memory HIV-1 specifc immune responses induced by vTTB8R/A52L-gpe and vTT-gpe. Results: Double deletion of B8R and A52L greatly reduced the virus replication in the brain of mice, but did not affect the outcome of intranasal infection of BALB/c mice with dose of 5104 or 5105 PFU. Flow cytometry analysis and ELISPOT revealed that both vTTB8R/A52L-gpe and vTT-gpe triggered HIV-1 specific CD8+ T cells, however, vTTB8R/A52L-gpe enhanced the magnitude of HIV-1 memory responses. Especially, vTTB8R/A52L-gpe reduced the vector-specific T cell response. Both vectors were capable of inducing similar levels of antibody against gp120. Conclusion: Double deletion of B8R and A52L could attenuate the neurovirulence in mice. Both vTTB8R/A52L-gpe and vTT-gpe induced robust T-cell response to HIV-1, but vTTB8R/A52Lgpe showed more durable HIV-1 memory responses and weaker vector-specific T cell response.

Background: Even though it has been more than twenty-five years since the discovery of HIV, an effective preventative vaccine remains elusive. The difficulty in developing an effective HIV vaccine using traditional approaches highlights the need for novel strategies. One novel vaccine strategy is to target the function of HIV-1 protease. Protease cleavage sites (PCS) of HIV-1 are highly conserved amongst the major subtypes. Proper cleavage of all 12 sites is vital in the generation of a viable virion. Directing immune responses against these cleavage sites could destroy the virus before it could establish itself in the host. Also, it would force the virus to accumulate mutations at the PCSs, thus eliminating the ability of the protease to generate infectious virions. In this study, we tested the feasibility of generating immune responses to the peptides corresponding to 12 PCSs of SIVmac239 using a vesicular stomatitis virus (VSV) vector. Methods: Thirty nucleotides upstream and downstream of each SIVmac239 PCS was codon optimized and cloned into pATXVSV-G. Each vaccine vector was rescued by transfection of 293T cells co-cultured with VEROE6 cells. PCS mRNA expression was confirmed by RT-PCR. BALB/c mice were used to test the expression of the peptides induced by recombinant VSVs (rVSVs). Each group was vaccinated (intramuscularly) with 1 of 12 purified recombinant VSVs (1x105 PFU). Control mice were injected with wild-type VSV. PCS peptide expression was confirmed using indirect enzyme-linked immunosorbent assay. Results: RT-PCR confirmed the expression of PCS mRNA by all 12 rVSVs, in vitro. Two and 4 weeks post-vaccination, peptide specific IgM was detected in all groups of mice immunized with the rVSVs, except mice vaccinated with rVSVs expressing PCS 7 epitope. Conclusion: We successfully generated 12 rVSVs expressing peptides overlapping the 12 PCS. Eleven of the 12 peptides were immunogenic in mice. Studies with non-human primates are currently underway.

AIDS Vaccine 2011

195

POSTERS

Posters
P18.11

Topic 18: T Cell Vaccine Concepts and Design P18.12


Assessment of the Feasibility of a Different HIV Vaccine Approach
R. Capina1, J. Tuff1, C. Daniuk1, D. La1, C. Czarnecki1, T. Bielawny1, H. Peters1, M. Kimani2, C. Wachihi2, J. Kimani2, B. Ball1, F.A. Plummer1, M. Luo1
1 2

Priming with a Mixture of Recombinant BCG Expressing HIV-1 Gag, RT and gp120 and Boosting with Recombinant MVA Induces an Effective Immune Response
R.E. Chapman1, E. Shephard1, H. Stutz1, G. Chege1, N. Douglass1, A. Williamson1
1

University of Cape Town, Cape Town, South Africa

Public Health Agency of Canada, Winnipeg, Canada; University of Nairobi, Nairobi, Kenya

Background: Mycobacterium bovis BCG (BCG) has a number of characteristics that give it great potential to act as a vehicle for the delivery of recombinant vaccines. However, its success depends on overcoming the challenges of poor antigen expression levels and genetic instability. Our studies using an optimized mycobacterial shuttle vector which utilizes the Mycobacterium tuberculosis mtrA promoter, induced upon infection of macrophages, and the M. tuberculosis 19 kDa signal sequence may overcome these issues. We have used this system to generate recombinant BCG (rBCG) expressing HIV-1 subtype C full length Gag, RT and gp120. The V3 CTL epitope, recognised by BALB/c mice, was translationally fused to the HIV proteins for immunogenicity assays. Methods: BALB/c mice were vaccinated with each rBCG vaccine alone or simultaneously with all three vaccines and boosted with a matched recombinant MVA (SAAVI MVA-C). Immune responses were monitored using an IFN- ELISPOT assay. Results: Mice primed with a single, simultaneous inoculation of all three recombinants and boosted with SAAVI MVA-C gave a peak V3 CTL-specific response of 708 73 spot forming units (SFU)/106 splenocytes. A high magnitude of RT-specific CD8+ T cells and a low magnitude of Gag-specific CD8+ and CD4+ T cells were also detected. Higher peak responses were observed to Gag and RT peptides in mice primed with a single rBCG than to those primed with a mixture of all three recombinants. Conclusion: These results show that a mixture of three recombinant BCG strains expressing HIV-1 Gag, RT and gp120 can prime a robust T cell response in mice when used in combination with a MVA boost. Acknowledgments: This work was supported by NIH, USA (grant# R21/R33-A1073182-01A1)

Background: The failure of Merck STEP and Phambili trials and the modest effect of RV144 trial emphasize the importance of understanding the correlates of protective immunity. Our study showed that the epitope recognition of HLA alleles associated with protection from HIV-1 infection is very narrow, thus vaccines focus on the key sites of HIV-1 might work better. Since the protease cleavage sites of HIV-1 are highly conserved among major subtypes, direct immune responses against these sites would yield two major advantages. First, the immune response could destroy the virus before it can establish permanently in the host. Second, the vaccine could force the virus to accumulate mutations eliminating the normal function of the HIV protease thus eliminating viable virions. For this vaccine strategy to work a given individual must have a HLA class I allele that can recognize one of the peptides overlapping one of the 12 protease cleavage sites of HIV-1. In this study we examined the population coverage of this vaccine approach using several approaches. Methods: The population coverage was predicted using computational algorithms, the Population Coverage Calculator (http://www.immuneepitope.org/) with the clade A and D peptides overlapping the protease cleavage sites (PCSs). The population coverage was also calculated based on the T cell epitopes that have already been identified at these sites. Furthermore, the peptides overlapping the 12 PCSs were screened with 8 HLA class I alleles using iTopia Epitope Discovery system and confirmed using IFN ELISPOT assays with PBMCs. Results: Analysis using all three approaches showed that the percentage of populations in the world can recognize peptides overlapping at least one PCS is very high, including more than 90% population in Sub-Saharan Africa. iTopia epitope Discovery System screen showed that the eight common HLA alleles have epitopes in multiple PCSs (4 to 12). Conclusion: This vaccine approach has good population coverage.

POSTERS 196 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.13


Immunogenicity of a DNA Vaccine Containing Multiple HLA-DR Binding, Conserved M Group HIV-1 Epitopes in BALB/c Mice
R.R. Almeida , S.P. Ribeiro , D.S. Rosa , J. Kalil , E. Cunha-Neto
1 1 1 1 1 1

Posters

P18.14
Using HLA Binding Prediction Algorithms for Epitope Mapping in HIV Vaccine Clinical Trials
D. He1, P. Kunwar2, E. Eskin1, H. Horton2, P. Gilbert3, T. Hertz3
1

University Of So Paulo, So Paulo, Brazil

University of California, Los Angeles, Los Angeles, CA, USA; Seattle BioMed, Seattle, WA, USA; 3Fred Hutchinson Cancer Research Center, Seattle, WA, USA
2

Background: An ideal T-cell based vaccine should elicit CD4 as well as CD8 T cell responses. The HIV-1 genetic diversity is a major concern in HIV vaccine development. Our group showed that a DNA vaccine encoding promiscuous multiple HLA-DR binding, conserved B-subtype HIV-1 CD4 T cell epitopes HIVBr18 induced broad cellular immune responses in mice. To include other prevalent HIV subtypes and increase the breadth of the induced response, we designed a new DNA vaccine containing a higher number of promiscuous epitopes, based on the conserved regions from HIV-1 M group consensus sequence. Methods: Regions of the HIV-1 M-group consensus sequence predicted to bind to 18 or more HLA-DR molecules tested by the TEPITOPE algorithm were selected. Among those, sequences with at least 50% conservation among HIV subtype consensus sequences were further selected. We identified 27 conserved, promiscuous HIV-1 CD4 T cell epitopes from 7 HIV-1 proteins and inserted their sequences in pVAX1. BALB/c mice were immunized with pVAX1, HIVBr18 or HIVBr27, 3x every 2 weeks. Immune responses were assessed 2 weeks after the last dose. Results: HIVBr27 and HIVBr18 elicited IFN- production in ELISPOT assays against 11 and 7 vaccine-encoded epitopes, respectively; similar results were observed in CD4+ and CD8+ proliferation assays. The magnitude of cytokine responses elicited by HIVBr27 was significantly higher than that induced by HIVBr18. HIVBr27 induced significantly higher frequencies of polyfunctional CD4+ and CD8+ T cells, able to proliferate and produce IFN- and/or TNF-, when compared to HIVBr18. Conclusion: The strategy of including a large number of promiscuous conserved M group epitopes in a DNA vaccine may thus be able to elicit a broad immune response against widely diverse HIV-1 isolates around the world, circumventing HIV-1 genetic diversity.

Background: A common endpoint in any T-cell based vaccine trial is measurements of vaccine-induced T-cell responses such as breadth and magnitude, measured using functional assays that measure the release of cytokines such as IFN-. The major limiting factor in these mapping studies is sample availability, as each test requires ~100K live cells. Current mapping strategies use a group-testing approach in which responses to an immunogen are measured using peptide pools that span a protein, and are then further refined using sets of mini-pools and a peptide matrix. Positive K-mers are then de-convoluted to identify the optimal epitope and restricting HLA allele. Methods: We explore the use of HLA binding predictors to improve the efficiency of epitope mapping protocols in vaccine trials. Given information about participants HLA alleles, we predict vaccine induced T-cell responses at various levels of refinement, based on the current group-testing hierarchical mapping approach. We benchmark these methods on epitope mapping data from a cohort of 12 HIV acutely infected individuals from the Seattle Primary Infection Cohort. Results: We find that incorporating HLA prediction methods provides significant improvements in mapping efficiency in each of the four stages of the current epitope mapping approach. For example, ranking all 9/10mer peptide-HLA pairs in a given reactive 15mer in order identify the optimal epitope, we find that 43/52 optimal epitopes were the highest ranked pair from a total of 66 potential pairs. We also find that by ranking the row and column mini-pools of a reactive peptide pool in order to identify the reactive 15mer, an average number of 9/20 minipools need to be tested, a savings of more than 50% of the number of tests. Conclusion: HLA binding predictors provide significant improvements to current mapping protocols and novel protocols that make use of these predictors should be designed and implemented.

AIDS Vaccine 2011

197

POSTERS

Posters
P18.15

Topic 18: T Cell Vaccine Concepts and Design P18.16


Determination of Cellular Immune Responses Induced by Consensus or Mosaic HIV-1 Env Vaccines in HLA-B7 Transgenic Mice
J. Yan2, T. Shin1, P. Pankhong1, A.S. Khan2, N.Y. Sardesai2, D.B. Weiner1
1

Characterization of Cell-Mediated Immune Responses Conferred by a Recombinant MeaslesHIV-1 Clade C Vaccine


R. Stebbings1, B. Li1, M. Fvrier2, C.M. Lorin3, M. Koutsoukos3, E. Mee1, N. Rose1, J. Hall1, M. Page1, F. Tangy2, G. Voss3, N. Almond1 National Institute for Biological Standards and Control (NIBSC), Potters Bar, United Kingdom (Great Britain); 2 Institut Pasteur, Paris, France; 3GlaxoSmithKline Biologicals, Rixensart, Belgium
1

University of Pennsylvania, Philadelphia, PA, USA; 2Inovio Pharmaceuticals, Philadelphia, PA, USA

Background: Live attenuated measles virus (MV) is one of the most efficient and safest vaccines available making it an attractive candidate vector for an HIV-1 vaccine aimed at eliciting cell-mediated immune responses. We first assessed the immunogenicity of a recombinant measles-HIV-1 vaccine (MV1-F4), carrying inserts for Gag, RT and Nef from HIV-1 Clade C, in mice (CD46/IFNAR). The vaccine candidate induced potent T cell responses with cross-reactive activity against Clade B antigens. Methods: We then characterized the immunogenicity of this vaccine candidate in cynomolgus macaques. Two groups of 8 MHC-matched macaques received intramuscular administration of Clade C MV1-F4 at days 0 and 84: group (A) received 104 CCID50 and group (B) 105 CCID50. Polyfunctional flow cytometry (CD3/CD4/ CD8/CD40L/IL-2/IFN/TNF) was performed to detect intracellular cytokine responses following stimulation with C-Clade synthetic peptide pools covering p24, RT, Nef and p17 sequences. Results: Significant HIV-insert-specific responses were detected in all individuals that received either the low dose or high dose of Clade C MV1-F4. CD4+ T cell responses to HIV-1 inserts were more frequent than CD8+ T cell responses. Boosting of insertspecific CD4+ T cell responses was observed after the second immunization in both groups, but was not observed for the CD8+ T cell responses. The intensity of the HIV-specific CD4+ T cell response was slightly increased when a high dose of the Clade C MV1-F4 vaccine was used as compared to low dose, but the levels of CD8+ T cells were comparable in both groups. The crossclade reactivity of the T cell responses and the presence of HIVspecific T cell responses in secondary lymphoid organ tissues are being investigated. Conclusion: Vaccination with Clade C MV1-F4 induces a robust and boostable cellular immune response in non-human primates, further supporting the exploration of the MV vector for HIV-1 vaccine strategies. Supported by the European Commissions Sixth Framework Programme (2005-019043 RMV-HIV).

Background: The genomic variability of HIV impedes development of globally relevant HIV vaccines. Developing vaccine platforms that induce strong Tcell breadth and magnitude has been difficult. In the past DNA vaccines were poorly immune potent in humans and were studied in prime boost approaches. Recently, data from the PENNVAX-B clinical trial (HVTN 080) showed that enhanced highly optimized DNA delivered by EP (E-DNA) drives strong CD4+ and CD8+ T cell responses that are equivalent/superior to viral & nonviral approaches. We build on these data by studying ways to further enhance the breadth and magnitude of E-DNA vaccines. Accordingly we compared two important epitope focusing strategies for their immune profiles, a novel optimized synthetic clade B env and a designed mosaic insert when both are enhanced for expression and delivered by EP. Methods: An optimized clade B consensus envelope vaccine (pEY2E1-B) and an optimized mosaic gp160 envelope vaccine were developed (pMosEnv). Following immunization with electroporation, cellular immune responses were assayed by IFN- ELISPOT in human HLA-B7 (mouse MHC KO) transgenic mice. Results: pEY2E1-B induced stronger cross-reactive responses against consensus peptides from individual subtypes compared to pMosEnv. The Elispot responses induced by pEY2E1-B against four pools of consensus subtype A, B and C peptides were 238, 1150 and 372. While the responses induced by pMosEnv were 54, 415 and 168. The consensus immunogen was up to 3x more potent at driving subtype-specific responses that crossrecognized diverse clade antigens. When a PTE peptide set was used, pMosEnv was more robust compared to pEY2E1-B. We have also explored prime boost approaches using these constructs and observed further robust immune enhancement. Conclusion: These strategies have unique strengths and specific differences in their immune targeting abilities. The consensus immunogen exhibits improved focus on conserved regions while the mosaic immunogen targets more variable epitopes. Combination approaches look very promising.

POSTERS 198 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.17


Design of Thermo-Stable Human/Simian Adenoviral T-Cell Based Vaccines Expressing Ubiquitin Fusions of Full or Fragmented HIV/SIV Genetic Components
T. Athanasopoulos1, A. Meiser2, V. Bachy3, A. Le Heron1, T. Papagatsias4, J. Harris1, A. Benlahrech2, S. Herath3, C. Hervouet3, P. Becker3, N. Roesen5, S. Oh6, F. Li6, S. Self7, S. Kwon6, K. Fischer5, L. Seymour5, R. Daniels8, M. Page9, N. Almond9, K. Logan2, R. Barbagalo2, L. Klavinskis3, S. Patterson2, G. Dickson1 Royal Holloway University of London, Egham, United Kingdom (Great Britain); 2Immunology Department, Imperial College London, London, United Kingdom (Great Britain); 3 Immunobiology, Kings College London School of Medicine, London, United Kingdom (Great Britain); 4Immunology Department, Imperial College London, United Kingdom (Great Britain); 5Genetics, Hybrid Therapeutics Ltd,, Oxford, United Kingdom (Great Britain); 6Development, TheraJect, Inc, USA; 7Vaccine Immunology Statistical Center-VISC, Fred Hutchinson Cancer Research Center, USA; 8VLP Development, The National Institute for Medical Research (NIMR), London, United Kingdom (Great Britain); 9 National Institute for Biological Standards and Controls (NIBSC), London, United Kingdom (Great Britain);
1

Posters

P18.18
DNA Vaccine Encoding HIV CD4 T Cell Epitopes Enhances CD8 T Cell Responses Induced by Immunization with DNA Vaccines Encoding Whole Gag (p55) and Vif
S.P. Ribeiro1, R.R. Almeida1, D.S. Rosa1, E.C. Mairena1, E.G. Kalls1, J. Kalil1, E. Cunha-Neto1 Faculdade de Medicinda a Universidade de So Paulo (FMUSP), So Paulo, Brazil
1

Background: Generation of competent and long-lived memory CD8+ T cells is highly dependent on CD4+ T cell help. A vaccine able to provide cognate help may efficiently prime CD8+ T cell responses to establish an appropriate immune control of HIV. Our group has identified a set of 18 conserved CD4 epitopes from 8 HIV-1 proteins. HIVBr18 DNA vaccine encoding such epitopes induced strong cellular immune responses in mice. Early Gag and Vif CD8+ T cell responses have been described as fundamental to control SIV replication. We aim test whether preimmunization with HIVBr18, encoding Vif and Gag CD4 epitopes, could boost Gag and Vif-specific CD8+ T cell responses induced by subsequent immunization with plasmids encoding whole Gag (p55) and Vif. Methods: Synthetic genes encoding the 18 HIV-1 previously identified CD4+ T cell epitopes or HIV-1 Gag and Vif whole sequences were subcloned in the pVAX1 vector to obtain HIVBr18, pVAX-gag and pVAX-vif, respectively. BALB/c mice were immunized with the plasmids pVAX-gag and pVAX-vif or preimmunized with the multiepitopic plasmid, HIVBr18, followed by immunizations with pVAX-gag and pVAX-vif, in a regimen of 4 doses. Results: Preimmunization with HIVBr18 elicited broader and stronger IFN-g secretion as well as T cell proliferation against Gag and Vif peptides than immunization with pVAX-gag + pVAX-vif alone. These increases were also observed against Gag and Vif regions not shared with the HIV CD4 epitope vaccine. Conclusion: Our results suggest that this vaccine may be useful as a source of cognate CD4+ T cell help for T cell responses, thereby increasing the efficacy of novel HIV-1 vaccine candidates. Financial Support. FAPESP, PN-DST AIDS -Ministry of Health, ICGEB, CNPq, INCT.

Background: Development, evaluation and optimisation of HIV-1/ SIV genetic vaccine components to generate a large pool of CD8+ T cell memory cells recognising multiple CTL epitopes, is currently one of the primary strategies in HIV-1/SIV vaccination protocols. Methods: We have identified sugar mixtures that give optimal preservation of adenovirus infectivity whilst maintaining the strength and rigidity of a micro-needle array. Results: Vectors retained substantial infectivity (i.e>55% eGFP+ cells) for up to 3 months through a temperature range (-20oC to +40oC). In vivo stability/infectivity studies have been successfully conducted via transcutaneous injections of dessicated Ad-OVAGFP vectors (4x10E9 vg) in C57BL/6 mice and footpad-mediated migration of Ad5-CMV-eGFP to lymph nodes. We originally aimed to induce immunity to HIV-1 infection by developing a cohort of adenoviral [human (Ad5/Ad11)/simian (Ad30/37)] vectors in which HIV-1 genetic components (gag/pol/env/nef) have been genetically engineered to stimulate broader CTL responses. As an example, full size HIVCN54 or SIVmac251 gag genes including an across clade conserved epitope were engineered, fused to mono-( N-end rule) or tetra-ubiquitin (Ub) sequences and further tested on DC and non-DC cell lines by using plasmid, in vitro synthesised mRNA, or human adenoviral vectors. We subsequently constructed a cohort (n=34) of rAd vectors carrying genetically fragmented ubiquitinated (4, 1, 0 Ub) fusions for either HIVCN54 (n=10) or SIVmac251 (n=7) gag genes attempting to reduce antigenic competition and/or alter epitope (sub)dominance. Conclusion: We have designed a cohort of human/simian adenoviral T-cell based vaccines expressing ubiquitin fusions of full or fragmented HIV/SIV genetic components. We are currently extending our studies using an in vitro CTL epitopemapping system employing human monocyte-derived DCs from healthy and HIV-1+ individuals alongside mouse in vivo studies impacting the antigen ubiquitination and genetic fragmentation on the quality of the generated immune responses. SIV constructs are currently forward tested in non-human primate (NHP) models (cynomolgus macaques).

AIDS Vaccine 2011

199

POSTERS

Posters
P18.19

Topic 18: T Cell Vaccine Concepts and Design P18.20


The GAIA HIV Vaccine Progress Report: Further Validation of Broadly Conserved Class I Epitopes in West Africa
L. Moise2, O. Koita1, K. Sangare1, L. Levitz2, J. Rozehnal3, Z. Koty4, K. Tounkara4, M. Ardito2, M. Rochas4, W. Martin2, A. De Groot5
1

Conservation Across Time and Sequence: Validation of Immunogenic HLA-A2 Achilles Heel Epitopes for the GAIA HIV Vaccine
K. Sangare1, O. Koita2, K. Tounkara1, C.M. Boyle3, L. Levitz3, J. Rozehnal4, Y. Kone1, Z. Koty1, M.A. Ardito3, W.D. Martin3, A. De Groot5
1

GAIA Vaccine Foundation, Bamako, Mali; 2LBMA-Laboratoire de Biologie Moleculaire Appliquee, Bamako, Mali; 3EpiVax Inc., Providence, RI, USA; 4Institute for Immunology and Informatics - University of Rhode Island, Providence, RI, USA; 5 EpiVax Inc.,GAIA Vaccine Foundation, URI, Brown Medical Department, Providence, RI, USA
Background: The genomic sequence variability of HIV has complicated efforts to generate an effective and globally relevant vaccine. Viral variation in response to humoral and cellular immune pressure not only alters established immune targets but may also have contributed to several recent HIV vaccine failures. HIV epitopes conserved in sequence, three-dimensional configuration and across time may represent regions of the viral genome that are constrained due to functional or structural limitations. These Achilles Heel epitopes of HIV would therefore be ideal candidates for inclusion in an epitope-based HIV vaccine Methods: In this study we used immunoinformatics tools to select highly conserved putative HLA-A2 epitopes. This analysis was first performed in 2002 on 10,803 HIV-1 sequences and reevaluated in 2009 on an expanded 43,822 sequences. Analysis done in 2009 revealed surprising stability of the 25 epitopes identified in 2002, and identified an additional 13 highly conserved HLA-A2 candidates. In total, 38 highly conserved HLA-A2 candidate epitopes were selected and evaluated for HLAbinding and ELISpot assays on PBMC from HIV infected patients in Providence, RI and/or Bamako, Mali. Results: Thirty-five (92%) of the 38 selected peptides stimulated IFN-g response in PBMC from at least one subject. Of the 25 peptides selected in 2002, 21 were validated by assays performed in Providence. Eleven (85%) of the 13 peptides selected in 2009 were confirmed assays performed in Mali. Twelve of 18 peptides assayed in both Providence and Mali were confirmed in both locations. Conclusion: Validation of these selected HLA-A2 epitopes conserved across time (2002-to-2009) and space (Providence and Mali), support the hypothesis that these epitopes would be appropriate candidates for inclusion in a globally relevant HIV vaccine.

Laboratory of Applied Molecular Biology and Faculty of Medicine, Bamako, Mali; 2EpiVax Inc., Providence, RI, USA; 3 Institute for Immunology and Informatics - University of Rhode Island, Providence, RI, USA; 4GAIA Vaccine Foundation, Bamako, Mali; 5EpiVax Inc.,GAIA Vaccine Foundation, URI, Brown Medical Department, Providence, RI, USA
Background: We set out to develop an epitope-driven, DNA-prime/ pseudoprotein-boost HIV vaccine (GAIA vaccine) composed of both CTL and T helper cell epitopes that are highly conserved and immunogenic across diverse HLA types and HIV strains, as well as over time and geography. The importance of generating broad T cell responses to protect against infection is reinforced by the successful outcome of the RV144 trial. Methods: 10,199 HIV protein sequences were searched for conserved 9-10-mer segments. From 5,494 of the most highly conserved 9-mer sequences (>5% isolates), HLA Class I binding sequences and Class II immunogenic consensus sequences (ICS) were identified for experimental validation and vaccine design using the EpiMatrix suite of immunoinformatic algorithms. Validations were performed in Thailand, the USA, and more recently Mali, by IFN-gamma ELISpot assay, using peripheral blood from HIV-infected donors. DNA-prime/peptide-boost vaccine studies including these epitopes were performed in HLA transgenic mice. Results: Individual epitopes selected for study are more broadly conserved than those chosen for other epitope-based vaccines (>70%, compared to Epimmunes 40%). Antigenicity of 98% of the ICS epitopes and Class I epitopes (87% A2, 29% A3, 67% B7, 20% A24) was confirmed in HIV-infected subjects. Fifteen ICS peptides and 12 A2 peptides were confirmed in both Mali and the USA. Notably, GAIA A2 epitope conservation across clades since 2002 is maintained with very little change, despite the expanding number of sequences available. Epitope immunogenicity was confirmed in HLA transgenic mouse immunization studies. Conclusion: The GAIA method for selecting HIV sequences identifies epitopes with broad coverage over time, HIV strains, and geographic regions for well-represented class I and II alleles in the general population. DNA vaccine prototypes encoding these epitopes are immunogenic in HLA transgenic mice. We anticipate the epitope-rich GAIA vaccine when tested in human volunteers will induce greater immunogenicity than have other DNA/viral vector prime-boost vaccines.

POSTERS 200 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.21 LB


A Phase IIB, Randomized, Double-Blind, Multicenter, Immunogenicity Study of Vacc-4x Versus Placebo in HIV-1-infected Patients
J.K. Rockstroh1, G. Pantaleo2, R. Pollard3, J. van Lunzen4, D. Asmuth3, B. Peters5, D. Podzamczer6, A. Lazzarin7, K. Ellefsen-Lavoie2, I. Baksaas8, M.A. Sommerfelt9, V. Wendel-Hansen9, B. Srensen9
1

Posters

P18.22 LB
The Improvement of Five HIV-1 B`/C Subtype Genes Modification on the Express Level In Vitro in HIV Vaccine
Y. Gao1, X. Qi1
1

National Insitute for Viral Diseases Control and Prevention, China CDC, Beijing, China

University of Bonn, Bonn, Germany; 2University of Lausanne, Lausanne, Switzerland; 3University of California Davis, Sacramento, CA, USA; 4University of Hamburg-Eppendorf, Hamburg, Germany; 5Guys and St Thomas Hospital, London, United Kingdom (Great Britain); 6Hospital Universitari Bellvitge, Barcelona, Spain; 7Vita-Salute San Raffaele University, Milan, Italy; 8Mericon AS, Skien, Norway; 9Bionor Pharma ASA, Skien, Norway
Background: Vacc-4x is a peptide-based HIV therapeutic vaccine to conserved domains on p24Gag. Recently conserved sectors on HIV p24, critical for virus viability and thereby immunologically vulnerable have been identified. Elite controllers target immune responses to such regions. The Vacc-4x peptides lie within a number of these conserved sectors of HIV p24. The co-primary endpoints of this study were to compare changes in CD4 counts and return to ART between treatment and placebo groups during a 24 week treatment interruption. Secondary endpoints included safety, viral load and immunogenicity Methods: This prospective, randomized, double blind phase IIB clinical study (NCT00659789) was carried out in 13 European and 5 US centers recruiting 135 patients on ART. After 6 immunizations on ART over 28 weeks, treatment was interrupted for up to 24 weeks (to week 52) (Vacc-4x n=88; placebo n=38). Immunological analyses (ELISPOT, proliferation, intracellular cytokine staining) were carried out at central laboratories.

Background: Since the first reported AIDS case in 1981, AIDS has been spreading at an alarming rate in the world, seriously threatened human health. HIV vaccine has not been developed yet in the past decades, because of the specific biological characteristics and the lack of awareness of the mechanism on HIV infection and immune protection. The neutralizing antibody induction is important for the development of traditional vaccine, but for the HIV vaccine, besides the induction of crossprotective neutralizing antibody, wide spectrum cellular response is also important for the high efficiency protection. It`s getting a important strategy for HIV cellular immune vaccine to modify cellular immune antigen, use multi-antigens and combine immumization of prime-boostwith different vaccines. Methods: we selected five major cellular immune antigens of HIV-1 B `/ C subtype as target genes, which are gag, pol, rev, tat and nef, to modify and optimize their gene sequences, codon bias and express structure, and then constructed DNA vaccines or recombinant vaccinia virus(rVV) to assess the express level of the genes in vitro, respectively. Results: Results shows that all optimized and non-optimized genes can express in DNA and rVV, and genes express better in DNA than rVV. Gag and pol had higher express level after gene optimization. The significant promote of express level is observed in pol gene, single pol gene had higher express level than gagpol fusing gene, but similar result was not observed in gag. As for rev, tat and nef, optimized single gene express slight higher level than optimized hRTN(fusing gene by rev,tat and nef), and both optimized single rev, tat, nef gene and fusing hRTN had better express level than non-optimized RTN. Conclusion: These data suggest that all the optimized HIV genes may be good vaccine candidate antigens for use as a protective HIV vaccine based on the cellular mechanism.

Results: There were no Vacc-4x-related serious adverse events. Of the 135 patients recruited (male n=92; female n=43), 126 patients completed the study. Median prestudy CD4 count was 712 (Vacc-4x) and 619 cells/mm3 (placebo), and median CD4 nadir 300 (Vacc-4x) and 285 cells/mm3 (placebo). There was no statistically significant difference between the two groups regarding change in CD4 counts (p=0.12) or ART resumption (p=0.89) during treatment interruption. A statistically significant treatment difference between Vacc-4x and placebo groups for viral load (VL) was found for patients who achieved a 6 month ART-free period (p=0.0022). There was a positive correlation between ELISPOT responses and lower viral load in the Vacc4x group compared to placebo (p=0.02). Long-term follow-up of patients up to week 104 was completed in June 2011. Conclusion: Vacc-4x was found to be safe and well tolerated. The Vacc-4x group experienced a significant reduction in viral load compared to placebo.

AIDS Vaccine 2011

201

POSTERS

Posters
P18.23 LB

Topic 18: T Cell Vaccine Concepts and Design P18.24 LB


Comprehensive Cross-Clade Elimination of Cells Infected with Globally Diverse HIV-1, HIV-2 and SIV Isolates by HERV-K-Specific CD8+ T-cells
B.R. Jones1, K.E. Garrison2, S. Mujib1, V. Mihajlovic1, N. Aidarus1, D.V. Hunter1, E. Martin1, V.M. John1, W. Zhan1, N.F. Faruk1, G. Gyenes1, N.C. Sheppard3, I.M. Pruimboom-Brees4, D. Goodwin4, D. SenGupta2, R. Tandon2, J.B. Sacha5, D.F. Nixon2, M.A. Ostrowski1 University of Toronto, Toronto, Canada; 2University of California, San Francisco, San Francisco, CA, USA; 3Vaccine Research, Worldwide Research and Development, Pfizer Inc, San Diego, CA, USA; 4Drug Safety RandD, Worldwide Research and Development, Pfizer Inc., Sandwich, United Kingdom (Great Britain); 5Oregon National Primate Center, Oregon Health and Science University, Beaverton, OR, USA
1

Fragmentation of SIV Gag induces Broader T cell Responses In Vitro


A. Benlahrech1, S. Herath1, A. Meiser1, T. Athanasopoulos2, F. Li3, S. Self3, P. Becker4, C. Hervouet4, L. Klavinskis4, G. Dickson2, S. Patterson1
2

Imperial College, London, United Kingdom (Great Britain); Royal Holloway University, Egham, United Kingdom (Great Britain); 3Statistical Center for HIV/AIDS Research and Prevention, Seattle, WA, USA; 4Kings College London, London, United Kingdom (Great Britain)
1

Background: Emergence of cytotoxic T lymphocyte (CTL) escape mutants during the natural course of SIV and HIV-1 infection suggests that CTL-exerted pressure plays an important role in the control of viral replication. Thus, broadening of the CTL response in a prophylactic setting is an attractive goal of T cell based vaccination against HIV-1. Antigenic competition is known to narrow the breadth of T cell responses. Therefore, we assessed whether gene fragmentation broadens the T cell repertoire. Methods: Ad5 vectors expressing SIVmac251 gag genes were used in this study. The vectors either expressed full length SIV-gag or one of 7 SIV-gag segments. These mini-genes were equal in size and neighbouring fragments overlapped by 10aa. Human dendritic cells (DC) were generated from monocytes and transduced with the different vectors separately. They were then co-cultured with autologous nave T cells that were isolated by negative selection. T cells were expanded over 3-4 weeks by boosting with autologous Ad5-transduced DC on a weekly basis. CD8 T cell responses against SIV overlapping peptides were then monitored by IFN- ELISPOTS. Their phenotype and ability to produce IFN-, TNF-, and IL-2 was also measured by flow cytometry. Results: No significant differences were observed between full length and fragmented SIV-gag constructs in terms of their ability to induce CD4 and CD8 T cell expansion. The generated cells exhibited an effector phenotype and had similar cytokine profiles in terms of IFN-, IL-2, and TNF-. However, full length SIVgag constructs generated CTL responses that recognised between 3 and 7 epitopes whereas fragmented SIV-gag vectors induced broader responses against new epitopes averaging 16 peptides. The magnitude of the response against most of the recognised epitopes was found to be higher when Ad5-vectors expressing fragmented genes were used. Conclusion: Antigen fragmentation augments the magnitude and breadth of CTL responses possibly through elimination of antigenic competition.

Background: The genetic diversity of HIV antigens represents a paramount challenge in the development of vaccines. Here, we establish rationale for circumventing this obstacle by eliciting cellular immune responses against stable human endogenous retrovirus K (HERV-K) antigens. Protein-level expression of HERV-K antigens has not been observed in healthy tissues, but does occur in various malignancies. We hypothesized that HIV would disrupt control of HERV-K expression within infected cells, targeting them for elimination by HERV-K-specific CD8+ T-cells. Methods: HERV-K expression was assessed by qPCR, western blot, immunohistochemistry and mass-spectrometry. HERVK-specific T-cell responses were identified by ELISPOT and a HERV-K-Env-specific CD8+ T-cell was cloned by IFN- capture. Additional HERV-K-specific T-cell lines and cloned were obtained by a dendritic-cell based expansion method. Recognition and elimination of HIV-infected cells were assessed by flow cytometry. Results: HIV-infection resulted in HERV-K Gag and Env protein expression in primary CD4+ T-cells. HERV-K Gag and Env specific CD8+ T-cells specifically responded to, and eliminated, HIVinfected cells in an MHC-I-restricted manner. This recognition was dependent upon Vif, however expression of Vif alone was insufficient to recapitulate a response. A HERV-K-Env-specific CD8+ T-cell clone displayed comprehensive elimination of cells infected with a panel of 23 diverse HIV-1 and HIV-2 primary isolates as well as with SIVmac251. Conclusion: The de-repression of HERV-K expression in HIVinfected cells, for which Vif is necessary but insufficient, constitutes a marker of infection that can be targeted by HERV-K-specific CD8+ T-cells. The unprecedented breadth of reactivity of HERV-Kspecific T-cell responses against diverse lentiviruses, both implied by the proposed mode of action and observed in the current study, comprises an enticing advantage over HIV-1-specific T-cell responses which could be exploited in the development of HERVK-targeted vaccines to treat or prevent HIV infection. These data also provides rationale for considering a role for HERV-K-specific T-cell responses in natural control of HIV.

POSTERS 202 AIDS Vaccine 2011

Topic 18: T Cell Vaccine Concepts and Design P18.25 LB


Robust Immunogenicity after HIV DNA Vaccination with IL-12 Plasmid Cytokine Adjuvant Delivered via Electroporation in HIV Uninfected Adults
S.A. Kalams1, S. Edupuganti2, M. Elizaga3, B. Metch3, J. Eldridge4, A. Sherwat5, S. De Rosa3, A.S. Khan6, N. Sardesai6, D.B. Weiner7
1

Posters

P18.26 LB
Immunological Correlates of Protection Against Acquisition of HIV-1 Infection in the iPrEx Trial
P.J. Kuebler1, S.J. Holditch1, J.J. Mcconnell2, L. Tarosso 3, M. Mehrotra 2, B.L. Shaw1, R.R. R. Andre1, C.G. Rex1, E.M. Eriksson 1, V.M. York1, V. McMahon2, R.J. Hance 2, P. Defechereux 2, D.V. Glidden2, S. Shiboski2, R.M. Grant2, D.F. Nixon1, E.G. Kallas3 University of California, San Francisco, San Francisco, CA, USA; 2J. D. Gladstone Institutes, University of California, San Francisco, San Francisco, CA, USA; 3Universidade So Paulo, Brazil
1

Vanderbilt University Medical Center, Nashville, TN, USA; Emory University School of Medicine, Atlanta, GA, USA; 3 Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 4 Profectus BioSciences, Inc., Tarrytown, NY, USA; 5National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA; 6Inovio Pharmaceuticals, Blue Bell, PA, USA; 7University of Pennsylvania, Philadelphia, PA, USA
2

Background: DNA vaccination in humans has demonstrated relatively poor levels of cellular immune responses. HVTN 080 tested the ability of HIV DNA combined with IL-12 plasmid, and delivered via electroporation (EP) to induce cellular immune responses in HIV-uninfected adults. Methods: HVTN 080 was a prospective, randomized, doubleblind, placebo-controlled trial of 48 healthy adults. Subjects received 3 vaccinations of PennVaxB (PV), which consisted of 1 mg each of HIV DNA Consensus B Gag, Pol and Env either alone (10 subjects) or in combination with 1 mg of GENEVAX IL-12 plasmid (30 subjects) at 0, 1, and 3 months via EP. Eight subjects received placebo. Pain was assessed via a 10-point visual analog scale immediately and at 5 and 25 minutes post-EP. Immune responses were measured by intracellular cytokine staining (ICS) assay. HVTN 070 evaluated 4 vaccinations (0, 1, 3, and 6 months) with 6 mg of PV and 1.5 mg of IL-12 plasmid in 30 subjects without EP. Results: The median pain score was 5.4 immediately, which decreased to 0.8 at 5 and 25 minutes. There were no significant vaccine-related adverse events. Two weeks after the 3rd vaccination 81% of PV + IL-12 subjects had a CD4+ T cell response, and 52% had a CD8+ T cell response. In contrast, response rates were significantly lower (CD4 responses 41%; p=.005 and CD8 T cell responses 4%; p=.002) when the same vaccine was given as 4 vaccinations at double the dose without EP (HVTN 070). Conclusion: HIV DNA vaccine+IL-12 plasmid delivered via EP led to frequencies of cellular immune responses equal or greater to those reported from current vector-based HIV vaccines such as adenovirus or traditional DNA vaccination without EP, either alone or followed by vector-based boost. Further trials delivering HIV DNA with or without IL-12 plasmid using EP as a vaccine strategy are merited.

Background: Pre-exposure prophylaxis for HIV-1 prevention in men who have sex with men (MSM) has been shown to reduce overall HIV-1 incidence by 44%. We hypothesized that exposure to HIV-1 before enrollment could have stimulated cell mediated immunity, which contributed to the observed protection. Methods: We studied immune responses in highly exposed MSM enrolled in the iPrEx study who either seroconverted (SC; N=93) or remained seronegative throughout follow up for up to 132 weeks (ESN; N=485). Pre-infection time-points from SC participants were matched with time-points from ESN controls and analyzed for HIV-1 specific T-cell responses to p24, Nef, Vif, integrase (Int), reverse transcriptase (RT), and protease (Prot) using a standard IFN ELIspot assay. Non-parametric tests were used to assess statistical significance of the comparisons between responses in SC and ESN participants. Results: ESN participants had a 3-fold greater mean aggregate HIV-1 specific T-cell response compared to SC participants, 297 vs. 99 SFU/106 PBMC, respectively (P<0.0001), and a greater breadth of responses (mean number of antigens recognized = 2.7 vs 1.6 (P<0.0001)). Conclusion: Broader and stronger HIV-1 specific T-cell responses in ESNs are a correlate of protection against acquisition of HIV-1.

AIDS Vaccine 2011

203

POSTERS

Posters
P18.27 LB

Topic 18: T Cell Vaccine Concepts and Design P18.28 LB


HVTN503/Phambili Vaccine Trial: The effect of the HIV-1 Clade B-Based Vaccine on Breakthrough Founder Viruses from a Clade C Infected Population
M. Logan1, T. Hertz2, P.T. Edlefsen2, A. DeCamp2, C. Magaret2, C. Rademeyer1, M. Rolland3, B.B. Larsen3, N. Frahm2, J. Marais1, R. Thebus1, F. Treurnicht1, H. Zhao4, j. Stoddard4, P. Konopa4, S. Nariya4, A. Lam4, J. Hural2, L. Corey2, J. Kublin2, G. Gray5, M.J. McElrath2, P. Gilbert2, J.I. Mullins4, C. Williamson1 University of Capetown, Capetown, South Africa; 2Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 3 Walter Reed Army Institute of Research, Silver Spring, MD, USA; 4University of Washington, Seattle, WA, USA; 5 University of Witwatersrand, Johannesburg, South Africa
1

HIV T-Cell Vaccines Induce Epitope Hotspots That Differ From Those Observed In Natural Infection and Target Variable Regions of the HIV Genome
T. Hertz1, N. Frahm1, H. Horton2, D. Friedrich1, L. Corey1, S. Buchbinder1, M.J. McElrath1, S.G. Self1, P. Gilbert1, N. HIV Vaccine Trial Network (HVTN)1
1 2

Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Seattle BioMed, Seattle, WA, USA

Background: Currently available T-cell based HIV vaccines induce detectable responses to a median of 1-2 epitopes. To analyze these vaccine effects, we generated and compared populationbased epitope maps from three T-cell HIV vaccine trials to those of natural infection. Methods: The vaccines used were Merck-Ad5 Gag/Pol/Nef (HVTN502/Step and Merck016) and VRC-Ad5 Gag/Pol/Env (HVTN054). We applied a group testing approach of IFN- ELISpot using overlapping peptides spanning the vaccine inserts and analyzed epitope mapping data from 71 HVTN502/Step, 72 Merck016 and 39 HVTN054 participants and 372 HIV-infected individuals. Epitope hotspots were defined as sites targeted more frequently than predicted by vaccine sequence alone. Statistical tests were developed both for hotspot existence and for hotspot comparison between vaccines. Results: We found strong evidence of multiple hotspots in all Step vaccine inserts: Gag, 4; Pol, 1; and Nef, 4 (p-values <0.00010.03). All vaccine inserts had at least one epitope hotspot. Several vaccine-induced hotspots were not present in natural infection or were specific to single vaccine trials. A statistical test measuring the significance of targeting frequency disparity between Merck and VRC vaccine-induced hotspots revealed differences for Gag and Pol (p=0.0035 and p=0.0004, respectively). Analysis of hotspot conservation patterns identified a bias toward conserved epitopes for HVTN054-induced (p=0.045) but not Step-induced (p=0.26) Gag hotspots. These data also showed Step Nef hotspots tended toward variable regions (p=0.002). Conclusion: To our knowledge, this is the first report to document the occurrence of immunodominant vaccine-induced hotspots from three different vaccine trials, several of which were not observed following natural HIV-1 infection. The finding that some hotspots target variable sites of the HIV genome highlights the need for future HIV vaccines that elicit more broad T-cell responses, notably to nonvariable sites. These findings have important implications for design of future vaccines and provide a simple framework for comparisons between different vaccine outcomes.

Background: The HVTN 503/Phambili trial (MRKAd5 gag/pol/nef HIV-1 subtype B vaccine) did not prevent infection nor reduce viral load set-point. While 80% of the vaccinees had insertmatched Gag-specific responses, only 50% recognized Gag subtype C PTE peptides. We characterized HIV-1 breakthrough infections from South Africans participants to determine if vaccination exerted a selective pressure on the infecting virus. Methods: 262 HIV-1 genome sequences were obtained from plasma at HIV-1 diagnosis from 20 placebo and 23 vaccine recipients. A pre-specified analysis plan was implemented to investigate possible vaccine sieve effects. Results: Of the 43 HIV-1 infections, 40 were subtype C and 3 were non-C; 24% vaccine and 26% placebo recipients were infected with heterogeneous founder populations. An average of 35% of 9mers in Nef founder sequences (12% in Gag) differed from the corresponding insert 9mer by > 2 amino acids. Unlike for the Step trial, for the analysis based on predicted epitopes, we found no significant difference in epitopic distances to the insert between vaccine and placebo group sequences. Analysis of sitespecific sieve effects revealed evidence for 3 signature sites: Gag 54, Gag 472 and Nef 23. A separate Bayesian analysis supported the findings at these sites (posterior probability > 0.999) and at several additional sites. Additionally, we found significantly different physio-chemical peptide properties at 9-mer beginning with Gag 471 (q-value = 0.14). Conclusion: Evidence for MRKAd5 sieve effects were previously demonstrated in the Step trial. Our results demonstrate much weaker MRKAd5 sieve effects in the Phambili trial, which may be explained by 1) few infection endpoints and thus limited statistical power; 2) incomplete vaccination courses; and 3) use of a subtype B immunogen in a predominantly subtype C infection. These findings underscore the importance of matching vaccine inserts to the predominant HIV-1 subtype of the region.

POSTERS 204 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.01


Enhanced DNA Vaccine with Protein Boost Delivered by EP Expands B and T Cell Responses and Neutralizing Phenotype In Vivo
K. Muthumani1, K. Broderick2, B. Bian1, A.A. Ramanathan1, P. Atman1, J. Yan2, X. Shen2, J. Mendoza2, A. Khan2, N. Sardesai2, D.B. Weiner1
1

Posters
P19.02
Design of a Dual Purpose Lentiviral Vector for Wild-Type CCR5 Phenotype Abolition, and CCR5 Delta-32/Delta-32 Genotype Transduction
M. Wayengera1
1

Unit of Genetics and Genomics, Kampala, Uganda

University of Pennsylvania School of Medicine, Philadelphia, PA, USA; 2Inovio Pharmaceuticals, Blue Bell, PA, USA

Background: The ability to elicit an improved level of antibody responses against HIV-1 is a crucial goal for a prophylactic HIV1 vaccine. DNA vaccines in the past have been poor inducers of both cellular as well as humoral immunity in humans. We have focused recently on the DNA immunogen design and Electroporation delivery to improve high levels of T cell immunity to HIV antigens in humans. Methods: Here we employ this advanced combination of technologies to focus on improving the B cell response induced by DNA vaccines to HIV plasmid encoded antigens. Furthermore we have compared this new DNA delivery to HIV envelop antigen vaccination as well as in a DNA prime + protein boost setting. Results: We observe for the first time that the antibody responses induced by this DNA platform alone are equal to and in some cases surpass the antibody responses induced by recombinant HIV env protein antigen in side by side studies. Furthermore in the prime boost setting both the quality and the magnitude of the antibodies are improved by up to several logs over protein immunization alone. We have also studied binding antibodies and observe that there is greater breadth of antibody binding induced by the DNA over that induced by protein, and this binding response is further enhanced in the prime boost setting. In addition the neutralization phenotype is expanded in the prime boost setting, over either immunogen platform alone to levels that mimic or surpass those induced by viral infection. Examination of T cell responses shows that the antibody enhancement does not negatively impact the high CTL responses generated by the DNA platform. Conclusion: This study suggests that these improved DNA construct designs and improved delivery are important for further investigation and optimization and may represent a uniquely malleable and important platform for HIV.

Background: Two chemokine receptors are known to participate in HIV entry: CCR5 and CXCR4. The CCR5 is coded by a gene located on chromosome 3, Locus 3p21.31, within the chemokine receptor gene cluster region. A naturally occurring CCR5 delta 32 mutant with a deletion in the second extracellular loop of the gene product results in impaired membrane expression of the receptor and leads to resistance to HIV-1 infection in homozygous individuals. There is evidence for the cure of HIV by transplantation of CCR5 delta 32/delta 32 stem cells. Stem cell transplantation is, however, not feasibly applicable for the cure of HIV world-wide. Affordable alternatives to CCR5 delta-32/delta-32 stem cell transplantation are required. Here, we advance a conceptual design of a single lentiviral vector for CCR5 delta-32/delta-32 genotype transduction. Methods: Design: Theoretical modeling; materials: CCR5 wildtype gene, CCR5 mutant gene, lentiviral vector design, and RNA interference model; Interventions: Three models were theoretically conceived: (a) wild-type CCR5 phenotype abolition, (b) mutant CCR5 (delta-32/delta-32) transduction, and (c) dual CCR5 wild-type phenotype abolition plus CCR5 mutant phenotype transduction. Results: We observed that- in CD4+ve progenitor cell-lines: First, expression of wild-type CCR5 phenotype can be abolished using a lentiviral vector carrying small interfering Ribonucleic acids (siRNA) targeted to any 18-22 linear base sequences within this genes delta-32 transcript region. Second, effective and efficient mutant CCR5 (delta-32/delta-32) transduction can recombinantly be achieved using a lentiviral vector carrying this traits gene sequence. Third, dual CCR5 wild-type phenotype abolition plus CCR5 delta-32/delta-32 phenotype transduction may be effected using a single lentiviral dual-purpose vector carrying (1) siRNA targeted to 18-22 linear base sequences within messenger RNA transcript corresponding to the 32 base-pair deletion, and (2) the entire CCR5 delta-32/delta-32 gene sequence. Conclusion: The dual-functional lentiviral vector modeled above offers a more affordable alternative for CCR5 delta-32/delta-32 genotype transplantation:- transduction.

AIDS Vaccine 2011

205

POSTERS

Posters
P19.03

Topic 19: Vaccine Concepts and Design P19.04


ENVolution: Immunoselection of Recombinant Vesicular Stomatitis Virus Expressing HIV1 Envelope Proteins by Broadly Neutralizing Antibodies
P. Tiberio1, M. Yuan1, K. Wright1, I. Lorenz1, S. Hoffenberg1, C. Jurgens1, C. Parks1
1

Purification of Native Env Trimers in Particulate and Soluble Forms and an Analysis of Their Stability and Antigenicity
T. Tong1, E. Crooks1, K. Osawa1, J. Binley1
1

Torrey Pines Institute for Molecular Studies, San Diego, CA, USA

Background: Native Env trimers may be ideal neutralizing antibody immunogens if they can be separated from antigenically promiscuous forms of Env that promote non-neutralizing responses. We recently found that enzyme digests substantially deplete junk Env from HIV particle surfaces, leaving native Env trimers intact. Here, we sought to: 1) Improve enzyme digest efficiency to produce pure trimer-VLPs 2) Evaluate trimer-VLP antigenicity 3) Isolate native Env trimers in soluble form 4) Evaluate the stability and antigenicity of soluble native trimers Methods: VLPs were glycosidase-protease digested and analyzed by blue native PAGE (BN-PAGE). Various digest conditions were modified to improve efficiency. The antigenicity of particulate trimers was assessed by VLP-ELISA and BN-PAGE. Soluble trimers were liberated from particles by detergent and their stability and antigenicity was assessed by BN-PAGE. Results: 1) Studies involving various Env mutants, the use of various enzymes and doses, and modifying incubation times and physical digest conditions led to substantial improvements in VLP digests, such that pure trimer VLPs can be isolated routinely. 2) In VLP-ELISA, undigested Env-VLPs were recognized by various mAbs, regardless of neutralizing activity. 2F5 bound substantially to particles lacking Env, presumably due to crossreactivity to membrane lipids. Digested wild type-VLPs (trimerVLPs)were recognized by neutralizing mAbs but not nonneutralizing mAbs. 3) Following VLP solubilization, SOS mutant trimers (disulfide linking gp120-gp41) could be isolated and remained stable. 4) Soluble SOS Env trimers were recognized only by neutralizing mAbs, including PG9 and PG16, suggesting the quaternary structure is intact. MPER mAbs bound more efficiently than they did to particulate trimers, perhaps due to a loss in membrane constraints. Conclusion: These results will enable the evaluation of pure native trimer immunogens in particle and soluble forms. Rabbit studies are ongoing. Our antigens could also assist in the isolation of new neutralizing mAbs and in determining the structure of native Env trimers.

International AIDS Vaccine Initiative, Brooklyn, NY, USA

Background: A formidable obstacle for human immunodeficiency virus (HIV) vaccine development is the design of an HIV envelope (Env) immunogen that elicits induction of broadly neutralizing antibodies (BnAbs), which block infectivity of several HIV strains. As with most RNA viruses, the vesicular stomatitis virus (VSV) RNA-dependent RNA polymerase lacks proof-reading function. Mutations accrue during serial passage for adaptation to specific environments. We have observed that recombinant VSV (rVSV) encoding a functional HIV Env in place of VSV G rapidly accumulated adaptive mutations in Env when propagated in the presence of BnAb b12 that enabled neutralization escape. This result suggests that selective pressure can be applied to rVSVEnv vectors to rapidly evolve novel HIV Env immunogens. BnAb b12 targets a discontinuous epitope near the CD4-binding site of gp120 subunit of HIV Env. The antigenicity of such epitopes may be altered by mutations that result in a conformational change of the overall trimeric complex; thus we currently are optimizing a system that employs VSVs evolutionary potential to generate novel Env glycoproteins selected based on their b12 binding properties. Methods: rVSV-GFP1-EnvG4 virus is captured by BnAb b12Protein G beads. Ribonucleoprotein (RNP) complexes of captured virus are extracted using detergent and salt. Purified RNPs are transfected into CD4/CCR5(+) cells to select out non-binders to b12 and enrich the population with only those viruses that retain b12 binding. Results: rVSV-GFP1-EnvG4 was immunoprecipitated by BnAb b12 as detected by Western Blot. Immunoprecipitated virus was successfully transfected into permissive cells after RNP extraction. Conclusion: A system has been established to select out HIV Env proteins based on binding to a BnAb. We will perform several rounds of this immunoselection coupled with serial passaging to examine if novel immunogens may be developed by this technology. This system may be applied to epitope mapping, immunogen design and neutralization escape.

POSTERS 206 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.05


Efficacy of DNA Vaccine Encoding RetrovirusBased VLP to Induce Potent Mucosal and Systemic Immune Responses and Its Use for HIV Vaccination
L. Torrieri-Dramard , F. Pitoiset , C. Huret , D. Desjardins , D. Klatzmann1, B. Bellier1
1 1 1 1 1

Posters
P19.06
Inherent Capacity to Develop Primary CTL Against Autologous HIV Epitope Variants: Implications for Prophylactic and Therapeutic Vaccines
K.N. Smith1, N.M. Melhem2, B. Colleton2, X. Huang2, W. Jiang2, R.B. Mailliard2, J.I. Mullins3, C.R. Rinaldo2
1

UPMC CNRS UMR7211, Paris, France

Background: The key properties of viruses that are responsible for eliciting potent immune responses may be used as a framework for rational vaccine design. These important immunogenic properties of viruses include their size, geometry, surface molecule organization and an ability to induce innate immunity with appropriate conditioning of the adaptive immune responses. New-generation vaccines aim to harness these properties. Methods: Consequently, we developed DNA-vaccines expressing recombinant retrovirus-based VLPs (plasmo-retroVLPs) pseudotyped with envelope glycoproteins expressed in their wild-type conformation. This strategy combines the efficiency of VLP-based vaccines with the simplicity and versatility of DNAbased vaccines. Results: Here, we demonstrate that plasmo-retroVLPs induce significantly better epitope-specific CTL and envelope-specific neutralizing responses than standard plasmids expressing nonparticulate antigens and thus constitute a new HIV vaccine candidate. We demonstrated in mice and macaques that plasmoretroVLPs elicit HIV-specific cellular and antibody immune responses. Furthermore, our results show that the delivery vehicle and route of immunization are critical factors governing the vaccine efficacy. Remarkably, we demonstrated that potent HIVspecific mucosal and systemic immune responses are induced after mucosal immunization using formulated DNA vectors. Conclusion: Taken together, DNA vaccines encoding HIVpseudotyped retroVLPs represent efficient immunogens that can be used to induce systemic and/or local immunity. Supported by CUTHIVAC (EU FP7 #241904) and ANRS.

University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; 2University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA; 3University of Washington School of Medicine, Seattle, WA, USA

Background: Dendritic cells (DC) have the unique ability to induce antiviral T cell immunity, making them ideal targets for HIV prophylactic and therapeutic vaccines. Here we use DC to demonstrate in vitro priming of HIV-specific CTL with effector function in HIV-negative donors. We then use this model to compare primary and memory T cell responses, using PBMC obtained before and after HIV infection, against autologous antigenic variants that accrued after infection. Methods: Epitope variants were revealed through viral sequencing, were evaluated for MHC binding, and used with Th1 polarizing DC to optimally prime CTL from HIV negative subjects. This DC-peptide model was then used to stimulate autologous T cells from pre-infection, autologous PBMC of HIV infected subjects in the Multicenter AIDS Cohort Study. Recall responses were examined by IFN ELISPOT using PBMC from multiple postinfection time points of these same subjects. Results: DC induced primary CTL against naturally evolving HLA A*0201 epitope variants in PBMC from HIV negative and HIV positive subjects, regardless of altered epitope binding to MHC. Despite loss of T cell recall responses during HIV infection to these variants, induction of primary CTL against these autologous epitopes demonstrated an inherent capacity to develop effector responses against naturally evolving virus variants. Conclusion: The ability to induce primary CTLs in vitro against autologous variant HIV epitopes in PBMC prior to seroconversion suggests healthy DC and T cells are able to overcome the burden of virus variation. The progressive loss in vivo of memory T cells against these variants suggests T cell dysfunction, and not solely viral mutation, is contributing to the inability to mount effective immunity during untreated infection. We therefore believe the immune system can effectively respond to virus variants and can be exploited to implement therapeutic vaccinations once T cell function is restored in HIV infected persons.

AIDS Vaccine 2011

207

POSTERS

Posters
P19.07

Topic 19: Vaccine Concepts and Design P19.08


Cutaneous and Mucosal HIV Vaccination EU-FP7 Funded Large-Scale Program
B. Combadiere1, B. Autran1, B. Bellier1, U. Blume-Peytavi2, C. Brander3, D. Costagliola4, I. Jani5, C. Katlama6, J. Sanchez7, S. MacCormack8, R. Shattock9, I. Stenescu10, B. Verrier11, A. Vogt2, R. Wagner12 UPMC INSERM U945, Paris, France; 2Charit Hospital, Berlin, Germany; 3IRSICAIXA, Barcelona, Spain; 4INSERM, Paris, France; 5INS, Mozambique; 6INSERM AP/HP, Paaris, France; 7 IMPACTA, Lima, Peru; 8MRC, London, United Kingdom (Great Britain); 9St Georges, University of London, London, United Kingdom (Great Britain); 10Fit Biotech, Finland; 11CNRS, Lyon, France; 12University of Resenburg, Resenburg, Germany
1

Refocusing the Antibody Response: The Use of an Antibody-Masking Technique to Elicit Broadly Neutralizing Antibodies
D. Wan , S. McMichael , G. Stewart-Jones
1 1 1 1

University of Oxford, Oxford, United Kingdom (Great Britain)

Background: The main goal of HIV-1 vaccine development is the design of an immunogen capable of inducing antibodies that can broadly neutralize HIV-1. However, it has proved to be extremely difficult to elicit such antibodies through vaccination as the host immune system is usually distracted by immunodominant regions on the HIV Env protein, such as the hypervariable loops, and the antibodies generated are non-neutralizing. Here, we describe an approach which we aim to refocus the immune response towards key epitopes as defined by known BNAbs by utilizing non-neutralizing antibodies as a masking shield of the immunodominant regions on HIV Env protein. Methods: Two immunogens have been designed aiming to encourage the elicitation of CD4bs-targeting and MPER-targeting antibodies in a two stage mice immunization schedule. All Env proteins are based on the Clade B HIV-1 JRFL. Female Balb/c mices (7 weeks old) are DNA primed followed by two protein boosts every three weeks. Sera were collected one week before and after DNA priming, and one week after each boost. IgG antibodies are purified from the pooled sera, and will be used to construct a masking shield in the second immunization. Results: The two immunogens were tested and verified against different BNAbs. IgG against these immunogens were purified and digested into their Fab region. Future works involve the use of purified IgG Fab to construct a masking shield against and in vitro testing of the masking mechanism. Immunization of mice using the shielded materials will be carried out should the initial testing proved promising. Conclusion: The masking of immunodominant regions on HIV Env protein by previously elicited non-neutralizing antibodies may prove to be a useful tool in refocusing the immune response towards key epitopes defined to date, and encourage the elicitation of broadly neutralizing antibodies.

Background: The CUTHIVAC project aims at assessing a new HIV vaccine strategy to prevent and control HIV infection based on transcutaneous and/or mucosal routes and immunogen delivery systems to preferentially promote and redirect immune responses towards high level of mucosal Abs and effector CD8 cell responses directed against various HIV antigens. Methods: Taking advantage of new genetic and immunological information provided by scientific, industrial and academic partners, CUTHIVAC will design, develop and validate innovative immunogens and delivery systems as well as immunization methods. Clinical trials will be implemented with the last cutting-edge generation of HIV DNA-GTU candidate applied by transcutaneous, intradermal routes and/or mucosal administration of HIV-envelope protein-based vaccine. Much work will be carried out on the new genetic design of HIV antigens and delivery systems for developed and developing countries. These new vaccines will be tested in innovative preclinical and innovative humanized mice approaches with a special highlight on routes of vaccination. The project also aims at rapidly translating preclinical approaches into prophylactic and therapeutic clinical trials in developed and developing countries that could help to prevent and eradicate HIV Results: The project will shed light on our understanding of the mechanisms of vaccine penetration in skin and mucosa, and will highlight their impact on the quality of immune responses. Based on strong scientific knowledge of HIV disease and new technical approaches in the field of vaccinology, CUTHIVAC will redesign efficient vaccine candidates to provide the basis for the introduction of entirely novel vaccination systems into the clinic. Conclusion: Through its integrative and multidisciplinary approach, CUTHIVAC will therefore provide the basis for a novel approach in vaccination with a view to widening its application to other infectious diseases such as malaria and tuberculosis.

POSTERS 208 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.09


Optimizing the Immune Response to MVA/HIV Vaccine Candidates in Mice
L. Wyatt1, W. Xiao1, P. Earl1, B. Moss1
1

Posters
P19.10
A Strategy to Circumvent Preexisting Adenovirus Neutralizing Antibodies Using Adenovirus-Infected Blood Cells: Proof-of-Concept in Rhesus Macaque
S. Caijun1, F. Liqiang1, Z. Linqi2, C. Ling1
1

LVD, National Institute of Allergy and Infectious Disease, National In, Bethesda, MD, USA

Background: Recombinant MVA expressing HIV envelope and gagpol genes (MVA/HIV) are currently being tested in phase I and II vaccine trials. In attempts to make MVA/HIV candidates more immunogenic, we have reassessed the immune response of mice immunized 1) by different routes, and 2) with different configurations of the HIV envelope (secreted gp140, membrane-bound gp160, trimeric HIV/SIV gp140 chimera) in prime-boost experiments. Methods: In one study, mice were immunized 2 times with MVA/HIV by intramuscular (IM), intradermal (ID), and skin scarification (SS) routes. In a separate study, mice were immunized by the SS route with viruses expressing HIV gp140, gp160, or trimeric HIV/SIV gp140 chimera and boosted with gp140 or gp140 chimera proteins. Vaccinia and HIV envelope CTL and binding antibody ELISAs, vaccinia and HIV neutralization assays (Clade B Tier 1 and Tier 2), and vaccinia challenge protection studies were performed. Results: Higher CTL responses to vaccinia and HIV envelope were found by immunization using IM and SS routes. Superior vaccinia and HIV envelope binding ELISA and vaccinia neutralization responses were seen using ID and SS routes. The SS route afforded the best protection against weight loss by intranasal lethal vaccinia challenge. In prime-boost experiments with different HIV envelope constructs, all 3 groups induced similar HIV envelope CTLs and binding ELISA antibodies. The gp160-immunized group induced the highest level of neutralizing antibodies with the order of the three groups being gp160>gp140>gp140 chimera. No increased breadth of neutralizing antibodies was detected. Conclusion: Taken together, our data suggest that MVA/ HIV immunization by the SS route produces optimal immune response, and that membrane-bound HIV envelope is superior to secreted gp140 in induction of HIV neutralizing antibodies. The trimeric structure of the HIV/SIV gp140 chimera did not increase the breadth of neutralizing antibody produced.

Guangzhou Institute and Biomedicine and Health, Guangzhou, China; 2Comprehensive AIDS Research Center, Tsinghua University, Beijing, China
Background: Adenoviral vectors have been extensively exploited for the development of prophylactic and therapeutic vaccines for a variety infectious diseases and cancers. However, the practical application of Ad5 could be limited by the high prevalence of preexisting anti-Ad5 immunity, especially Ad5 neutralizing antibodies. Methods: In the present study, we found that freshly isolated PBMCs, mostly CD14+ cells, from Ad5-seropositive primates (humans and rhesus macaques) can be infected with Ad5 vectors. Interestingly, these cells are infected more efficiently than those from Ad5-seronegative individuals. On the basis of this observation, a novel strategy to circumvent the attenuated delivery efficiency due to Ad5 neutralizing antibodies was explored. Results: We demonstrated that the infusion of autologous PBMCs infected in vitro with Ad5-SIVenv can elicit robust immune responses in Ad5-seropositive rhesus macaques. Repeated infusion of Ad5-SIV vaccines carrying either homologous or heterogonous SIV antigens using this strategy generated antigenspecific immune responses in those monkeys. Furthermore, the monkeys were challenged intravenously with highly pathogenic SIVmac239 virus, and this strategy resulted in a significant reduction of the viral load at both the peak time and at a setpoint period compared with control animals. Conclusion: The results of this study warrant the further development of the adenoviral vector-infected PBMCs (AVIP) strategy as a simple but practically effective method for repeated delivery of Ad5-based vaccines.

AIDS Vaccine 2011

209

POSTERS

Posters
P19.11

Topic 19: Vaccine Concepts and Design P19.12


A Novel Vaccine Design for Inducing Robust HIV-1 Specific Immunity
1

Novel Non-Integrative One-Cycle Lentiral Genomes Derived from a Naturally Attenuated Animal Lentivirus as HIV Vaccines
G. Arrode-Brusses , D. Aldebert , B. Ouzrout , A. Smaoune , M. Moussa1, Y. Chebloune1
1 2 1

Z. Chen1, J. Zhou1, H. Wang1, Z. Tan1, H. Liu1, X. Lu1, Y. Kang1, Y. Du1, L. Liu1, Y. Xu2, L. Chen2 AIDS Institute, The University of Hong Kong LKS Faculty of Medicine, Hong Kong, China; 2Shanghai Teresa Healthcare Sci-Tech Co. Ltd,, Shanghai, China
1

INRA, UMR5163 CNRS-UJF, Grenoble, France; UMR5163 CNRS-UJF, Grenoble, France


1 2

Background: A safe and efficacious HIV vaccine is strongly needed to stop the continuing epidemics in humans. We developed lentiviral-based vectors as HIV DNA vaccines and have shown that our first generation of vectors was efficient alone to induce unique T cell responses and protection of vaccinated macaques from AIDS against pathogenic virus. We engineered a novel onecycle replication, non-integrative (NONI-LV) lentivector driven by lentiviral LTRs that have constitutive promoters. NONI-LV was used in the NOD/SCID-hu mouse model to examine HIV-specific immune responses. Methods: SIV LTRs were replaced with CAEV LTRs in the genome of SHIV-KU2 and the integrase gene was deleted. Resulting NONI-LV vaccine genome was transfected into HEK-293 T cells and expressed viral proteins were detected by RIPA analysis. Virus in the culture medium was examined for its capacity to induce a single cycle of infection in CEM-T4 cells. BALB/c and PBMC-humanized NOD/SCID beta2 mice were immunized with the NONI-LV DNA vaccine and humoral T cell immune responses were examined by ELISA and IFN-g ELISPOT. Results: RIPA analysis of viral proteins showed classical profiles in samples of cells transfected with NONI-LV and SHIV-KU2 DNAs. Infection of CEM-T4 cells showed a single round of infection of the culture medium from NONI-LV transfected cells, in contrast samples from SHIV-transfected cells produced virus that causes productive infections. NONI-LV and SHIV-KU2 DNAs but not D4SHIV-KU2 immunized mice induced HIV-specific antibody responses in sera of NOD/SCID-hu immunized mice. All NOD/ SCID-hu mice immunized with the NONI-LV developed potent T cell IFN-g ELISPOT responses that were nearly equivalent to those induced by SHIV-KU2 and much higher than those induced by D4SHIV-KU2 DNAs. Conclusion: These data show a substantial increase of immunogenicity of the new vaccine compared to our former DNA vaccine. Ongoing experiments will help to characterize the induced responses and to examine the correlates of protection.

Background: An ideal AIDS vaccine should induce broadly protective neutralizing antibody and cellular immune responses to cover all HIV-1 subtypes. To date, conventional vaccine strategies have failed to achieve this goal. It is, therefore, necessary to explore novel vaccine concepts for AIDS vaccine design. We hypothesized that a novel vaccine design might be targeting the test antigen to dendritic cells more effectively while inhibiting the negative effects of the PD-1/PD-L pathway on T cell function. Methods: We investigated a novel DNA vaccine design by fusing a test antigen, human HIV-1 Gag p24, with the soluble functional domain of programmed death 1 (sPD-1). This sPD-1-p24-DNA and control vaccines (e.g. mutant-sPD-1-p24-DNA, lacking the ligand binding domain) were used to vaccinate groups of BALB/c mice via intramuscular electroporation (EP). Vaccinated animals were subsequently sacrificed for measurement of HIV-1 Gagspecific responses using ELIspot, ICS, tetramer and ELISA assays. Vaccinated mice were also challenged with a vaccinia-Gag virus for efficacy evaluation. Results: Purified sPD-1-p24 interacted specifically with cells expressing ligands PD-L1 and PD-L2, as well as with bonemarrow derived primary dendritic cells. sPD-1-p24-DNA/ EP induced significantly higher levels of Gag-specific IFN- releasing CD8+ and CD4+ T cells determined by ELISpot assay. These specific T cells were polyfunctional, mainly co-expressing IFN- and TNF-, accounting for up to 27% and 4% of total CD8+ and CD4+ T cell populations, respectively. Moreover, up to 22% of total CD8+ T cells stained positive for HIV-1 Gagtetramer. Greater anti-Gag antibody titer was also observed with likely balanced IgG1 (Th2) and IgG2a (Th1) responses. Importantly, strong cell-mediated and antibody responses lasted over seven months and sPD-1-p24-DNA/EP-induced immunity provided significant protection against intranasal vaccinia-gag viral challenges. Conclusion: We have identified a novel vaccine design for inducing robust HIV-1 specific immunity, which warrants further investigation as an AIDS vaccine strategy. (We thank HKU-UDF and HKRGC762209 for support).

POSTERS 210 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.13


Immunogenicity of LC16m8 Vaccinia Virus/ Sendai Virus Vector Expressing the gp160 of HIV1 and Effect of CD40Lm
H. Shida1
1

Posters
P19.14
Mucosally Applied HIV gp140 DNA Prime/Protein Boost Strategies Generate Strong Serum and Mucosal Antigen-Specific Humoral Responses in Mice
J.F. Mann1, P.F. McKay1, R.K. Patel2, I. Vallado1, J.S. Tregoning1, R.J. Shattock1
2

Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan

Background: To develop an effective anti HIV-1 vaccine, elicitation of both humoral and cell-mediated immune responses is required. Previously we constructed a novel vaccinia virus (VV) strain, named LC16m8 that is replication competent but still safe in humans particularly because of its very low neurovirulency. LC16m8 rendered mice 1000 fold more resistant to pathogenic VV WR strain than nonreplicating VV MVA did. It is our aim to develop vaccination regimes that would efficiently induce both anti-HIV-1 humoral and cell-mediated immunities based on LC16m8. Methods: We tested various vaccination regimes including HIV-1 gp160 expressing plasmid prime followed by gp160 expressing LC16m8 (m8gp160) boost, and the m8gp160 prime followed by gp160 expressing Sendai virus (SeVgp160). Moreover we examined the adjuvant effect of a noncleavable mutant of human CD40L (CD40Lm) in C57BL/6 mice. Then we analyzed cellular immunities by ICS and antibodies using ELISA and Envpseudotyped viruses/TZM-bl cells. Results: Prime-boost immunization with plasmid and m8gp160 efficiently elicited Env specific IFN--producing CD8+ T cells but not any anti Env antibodies (Abs). However, the primeboost regimen using m8gp160 and SeVgp160 induced both IFN--producing CD8+ T-cells and neutralizing anti HIV-1 Abs. Moreover, CD40Lm expressing m8 co-immunization with m8gp160 elicited more CD8+ T-cell as well as anti Env Abs. Conclusion: These observations suggest that LC16m8 prime/ SeV boost regimen and CD40Lm expressing LC16m8 could be a vaccine eliciting both potent cellular and humoral immunities against HIV-1.

Imperial College, London, United Kingdom (Great Britain); St. Georges, University of London, London, United Kingdom (Great Britain)
1

Background: Mucosal surfaces are the first line of defence against most pathogens but there remains a distinct lack of mucosally applied vaccines capable of eliciting potent and effective mucosal immunity. We therefore aimed to investigate the utility of mucosally applied plasmid DNA as a means to initiate both systemic and mucosal humoral responses, which could then be greatly potentiated by protein boosting. Methods: Plasmid DNA was complexed to PEI and administered in a 15ul volume to either vaginal, sublingual or nasal mucosa of medroxyprogesterone treated BALB/c mice. Immunisations occurred fortnightly and mice were sampled (serum and vaginal wash) 7 days post each immunisation. Antigen-specific IgG and IgA production was assessed in the sera and mucosal lavage samples by quantitative ELISA and by splenocyte B cell ELISpot. Results: DNA topically applied to all mucosal surfaces tested primed gp140 antigen-specific humoral responses. Nasal and sublingual immunisation elicited antigen-specific IgG in serum and vaginal lavage however the latter was unable to generate mucosal IgA. Furthermore, both groups developed IgG and IgA spot forming units in splenocyte cultures. Vaginal immunisation generated low antigen-specific IgA antibody responses in serum and mucosal lavage in the absence of any IgG or B cell responses in the spleen. Finally, the nasal application of a heterologous DNA prime and gp140 protein boost revealed a single DNA prime to be sufficient to generate strong systemic and mucosal responses. Conclusion: We have performed a series of linked studies in the murine model that aimed to enhance vaccination via the mucosal route and to determine the potential role of plasmid DNA in mucosal immunisation regimens. Here we conclude that plasmid DNA encoding a large transgene product can be complexed to PEI and applied to various mucosal surfaces in a small volume formulation and that ensuing humoral immune responses vary depending on the administration site.

AIDS Vaccine 2011

211

POSTERS

Posters
P19.15

Topic 19: Vaccine Concepts and Design P19.16


Priming with Synthetic Nanovaccine Followed by rMVA Boost Is the Best Combination to Elicit Strong and Long-Lasting Gag or Env Immune Responses
S. Munier5, P. Mercier5, A. Calamel5, N. Bosquet1, T. Delair2, S. Paul3, P. Earl4, R. Le Grand1, B. Verrier5 CEA, Fontenay aux Roses, France; 2University of Lyon, Lyon, France; 3University of Saint Etienne, Saint Etienne, France; 4 National Institutes of Health, Bethesda, MD, USA; 5Centre National de la Recherche Scientifique, Lyon, France
1

HIV-Gag VLPs Presenting Trimeric HIV-1 gp140 Spikes Constitutively Expressed in Stable Double Transfected Insect Cell Line
M. Tagliamonte2, M. Visciano2, M. Tornesello2, A. De Stradis1, F.M. Buonaguro2, L. Buonaguro2
1

National Research Council, Institute of Plant Virology, Bari, Italy; 2National Cancer Institute, Naples, Italy

Background: We have previously described the establishment and characterization of a stably transfected insect cell line for the constitutive and efficient expression of Pr55 HIV Gag proteins, which auto-assemble into enveloped Virus-Like Particles (VLPs) released into the cell culture supernatant. Such HIV-Gag VLPs have been shown to elicit a specific systemic humoral response in vivo, proving the appropriate antigenic presentation of the HIV Gag protein to the immune system. Methods: Here we describe the establishment of a stable double transfected insect cell line for the constitutive and reproducible production of Pr55Gag-VLPs expressing on their surface trimeric forms of HIV-1 envelope glycoproteins. Results: The persistence of HIV coding genes has been verified in clonal resistant insect cells, the protein expression and conformation has been verified by Western Blot analysis. The resulting HIV-VLPs have been visualized by standard transmission electron microscopy and their immunogenicity has been evaluated in vivo. Conclusion: This represents, to our knowledge, the first example of stably transfected insect cell line for the constitutive production of enveloped HIV-Gag VLPs presenting trimeric HIV-gp140 on their surface.

Background: Heterologous prime boost vaccination is the best strategy to induce broad and potent immune responses. As the best combination implies the use of viral vectors, pre-existing immunity preclude their use on a repetitive basis. Recent advances in HIV biodegradable nanovaccines technology permit to investigate innovative prime boost strategy by combining synthetic and viral vectors Methods: We designed Poly(Lactic Acid) (PLA) nanoparticles (200 nm) carrying HIV-1 or SIV antigens as synthetic vectors, and rMVA or Ad5 as viral vectors. Different groups of mice were primed by subcutaneous route with viral vectors and boost with nanoparticles or vice/versa. Combination efficacy was assessed using p24 IgG,IgAs dosage and CTLs,Elispot assays. Best combination has been further evaluated in cynomolgus using PLA nanoparticles in presence of Poly I:C, followed by rMVA boost. Results: When considering the broadest immune response, best combination was a double priming with PLAp24 nanoparticles followed by viral vector, either Ad5gag or rMVAgag. When using opposite order, cellular and humoral immune responses was lower. Interestingly enough, vaginal IgGs were also observed in mice with a PLA prime/rMVA boost, and vaginal or rectal IgAs could be significantly increased after intranasal delivery of such combination. In cynomolgus experiment, three prime of PLA nanoparticles carrying SIV p27 or HIVgp140 in presence of poly I:C, followed by one boost of rMVASIVgag or rMVA gp140 CladeC, was able to induce strong and long lasting humoral immune responses against both antigens in sera, up to one year. Analysis of mucosal and cellular immune responses is currently being performed. Conclusion: We described for the first time a new heterologous prime/boost strategy using synthetic biodegradable vector as a prime and viral vector as a boost. Unexpectedly, priming with synthetic vector followed by viral boost, is the best strategy for inducing broad and long lasting HIV immune responses.

POSTERS 212 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.17


Immunogenicity and Efficacy of a Heterologous Mucosal Prime Intramuscular Boost Vaccination Strategy Against SIVmac239 Challenging
L. Zhang1, C. Sun2, Y. Du3, L. Chen2, Z. Chen3 Comprehensive AIDS Research Center, Tsinghua University, Beijing, China; 2Guangzhou Institute of Biomedicine and Health, CAS, Guangzhou, China; 3AIDS Institute of The University of Hong Kong, Hong Kong, China
1

Posters
P19.18
A Candidate HIV/AIDS Vaccine (MVA-B) that Enhances the Magnitude and Polyfunctionality of Memory HIV-1-Specific T-Cell Responses
J. Garca-Arriaza1, J. Njera1, C.E. Gmez1, N. Tewabe1, C.S. Sorzano2, T. Calandra3, T. Roger3, M. Esteban1 Centro Nacional de Biotecnologa, CSIC Madrid, Spain; Biocomputing Unit, Centro Nacional de Biotecnologa, CSIC, Madrid, Spain; 3Infectious Diseases Service, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
1 2

Background: Conventional vaccination strategies have failed to induce broadly reactive neutralizing antibody and cellular immune responses to prevent HIV-1 infection. It is, therefore, critical to test novel strategies in an efficacy study. Methods: We have generated a recombinant replication-competent modified vaccinia Tiantan (MVTT), namely rMVTTSIVgpe, as a mucosal vaccine expressing SIVmac239 Gag, Pol and Env. The immunogenicity and efficacy of rMVTTSIVgpe was studied in combination with an rAd5-based vaccine rAd5SIVgpe in Chinese macaques (Macaca mulatta) without the protective MHC class I allele Mamu-A*01. rMVTTSIVgpe was given through intranasal and oral inoculations whereas rAd5SIVgpe was through intramuscular injection. Four macaques in each of the four study groups received following prime and boost vaccinations: rMVTTSIVgpe/rAd5SIVgpe; rMVTTSIVgpe/rAd5SIVgpe, twice; rAd5SIVgpe/rAd5SIVgpe and placebo controls, respectively. Results: The heterologous rMVTTSIVgpe/rAd5SIVgpe regimen elicited cellular immune responses with significantly enhanced magnitude, breadth, sustainability, and poly-functionality when compared with the homologous rAd5SIVgpe regimen. Higher levels of neutralizing antibody (Nab) responses were also induced by the rMVTTSIVgpe/rAd5SIVgpe regimen against the sensitive SIVmac1A11 strain. After intrarectal challenge with a pathogenic and Chinese macaque-adapted SIVmac239 (5x105 TCID50 per animal), one of four monkeys vaccinated with the rMVTTSIVgpe/ rAd5SIVgpe regimen was fully protected whereas the rest showed an 1.5-1.9 log and 1.8-2.2 log reduction of peak and set-point viral loads as compared with control animals, which was reproducible in a separate study using additional four macaques. The control of viremia was likely correlated to CMI against Gag and Pol. 586 days post viral challenge all four monkeys vaccinated with the rMVTTSIVgpe/rAd5SIVgpe regimen remain clinically healthy whereas 2/4-3/4 animals in other groups have died of SAIDS. Conclusion: These data demonstrated that one-time rMVTTSIVgpe/ rAd5SIVgpe regimen induced durable immune control of a pathogenic, neutralization-resistant SIVmac239 challenge. Our findings have critical implications for the development of effective vaccination strategies against HIV-1 by engaging the mucosal immune system.

Background: The poxvirus vector Modified Vaccinia Virus Ankara (MVA) expressing HIV-1 Env, Gag, Pol and Nef antigens from clade B (MVA-B) is currently used as a HIV/AIDS vaccine candidate. A general strategy to try to improve the immunogenicity of poxvirus HIV-1 vaccine candidates is the deletion of known or suggested immunomodulatory vaccinia virus (VACV) genes. Methods: We have generated and characterized the innate immune sensing and the immunogenicity profile of a new HIV-1 vaccine candidate, which contains a deletion in a VACV gene. Results: We show that this VACV protein is expressed early during virus infection and localizes to the cytoplasm of infected cells. Deletion of this VACV gene from the MVA-B had no effect on virus growth kinetics; therefore this VACV protein is not essential for virus replication. The innate immune signals elicited by the MVA-B deletion mutant in human macrophages and monocyte-derived dendritic cells were characterized. In a DNA prime/MVA boost immunization protocol in mice, flow cytometry analysis revealed that the MVA-B deletion mutant enhanced the magnitude and polyfunctionality of the HIV-1-specific CD4+ and CD8+ T-cell memory immune responses, with most of the HIV-1 responses mediated by the CD8+ T-cell compartment with an effector phenotype. Significantly, while MVA-B induced preferentially Env- and Gag-specific CD8+ T-cell responses, the MVA-B deletion mutant induced more GPN-specific CD8+ T-cell responses. Furthermore, the MVA-B deletion mutant enhanced the levels of antibodies against Env in comparison with MVA-B. Conclusion: These findings revealed that this new VACV protein can be considered as an immunomodulator and that deleting this gene in MVA-B confers an immunological benefit by inducing innate immune responses and increasing the magnitude and quality of the T-cell memory immune responses to HIV-1 antigens. Our observations are relevant for the improvement of MVA vectors as HIV-1 vaccines.

AIDS Vaccine 2011

213

POSTERS

Posters
P19.19

Topic 19: Vaccine Concepts and Design P19.20


Impact of Pre-Existing Immunity on the Selection of Rare Adenovirus Vector Candidates: Implications for HIV Vaccine Development
M. Perreau1, H.C. Welles1, A. Harari1, L. Potin1, R. Martin1, S. Bibert2, T. Roger2, T. Calandra2, A. Bett3, J. Gall4, E.J. Kremer5, G. Pantaleo1
1 Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; 2Centre Universitaire Hospitalier Vaudois/ Infectious Diseases, Lausanne, Switzerland; 3Merck Research Laboratories, West Point, PA, USA; 44GenVec Inc., Gaithersburg, MD, USA; 5Institut de Gntique Molculaire de Montpellier, Montpellier, France

HIV CN54gp140 + GLA Significantly Enhances Vaccine Antigen-Specific T and B Cell Immune Responses After Priming with DNA and MVA
P. McKay1, A. Cope2, J. Swales2, S. Joseph3, M. Esteban4, R. Tatoud2, D. Carter5, J. Weber2, R.J. Shattock2
1

St. Georges, University of London, London, United Kingdom (Great Britain); 2Imperial College, London, United Kingdom (Great Britain); 3MRC Clinical Trials Unit, London, United Kingdom (Great Britain); 4Centro Nacional de Biotecnologa, CSIC, Madrid, Spain; 5Infectious Disease Research Institute, Seattle, WA, USA
Background: Using a unique vaccine antigen matched and single Clade C approach we have assessed the immunogenicity of a DNA-poxvirus-protein regimen in mice, administering the MVA and protein either sequentially or simultaneously with a novel TLR4 adjuvant (GLA). Methods: Groups of 10 BALB/c mice were primed with plasmid DNA encoding CN54 env/gag-pol-nef and then boosted with MVA-C (env-gag-pol-nef) and CN54gp140 protein with or without GLA (aqueous formulation). The MVA and protein were either given sequentially at 3 weekly intervals or simultaneously in different legs at 3 and 6 weeks. Mice were sampled (serum and vaginal wash) prior to each vaccination and three weeks after the final immunization. Absolute levels of antigen-specific IgG and IgA were measured in sera and mucosal lavage samples by quantitative ELISA. Splenocytes were harvested at necropsy and analysed for antigen-specific T cell responses using peptide pools spanning the Env and Gag proteins by IFN-gamma ELISpot assay. Results: GLA-adjuvanted CN54gp140 substantially enhanced the antigen-specific antibody responses in animals primed with DNA and MVA or MVA alone. Administration of MVA and CN54gp140 protein at the same time did not significantly affect antigen-specific antibody responses. Importantly, however, such co-administration of MVA-C and adjuvanted CN54gp140 did significantly augment antigen-specific T cell responses to Gag peptide pools. Conclusion: We have shown that GLA adjuvanted CN54gp140 is able to significantly boost vaccine antigen antibody responses and have further demonstrated that co-administration of the MVA and adjuvanted protein was equally effective to a sequential vaccination modality. This vaccine schedule shortens the duration of and simplifies the immunization regime, both central to long-term vaccine feasibility. In addition, a significant benefit of the combined inoculation was that T cell responses to proteins expressed by the MVA were potently enhanced, an effect that was likely due to the presence of systemic GLA.

Background: Adenovirus serotype 5 (Ad5) phase IIb vaccine trial (STEP) was prematurely stopped due to a lack of efficacy and twofold higher incidence of HIV infection among Ad5 seropositive vaccine recipients. We have recently demonstrated that Ad5 immune complexes (Ad5 ICs)-mediated activation of the dendritic cell (DC)-T cell axis was associated with the enhancement of HIV infection in vitro. Although the direct role of Ad5 neutralizing antibodies (NAbs) in the increase of HIV susceptibility during the STEP trial is still under debate, vector-specific NAbs remain a major hurdle for vector-based gene therapies or vaccine strategies. To surmount this obstacle, vectors based on rare Ad serotypes including Ad6, Ad26, Ad36 and Ad41 were engineered. Methods: The present study aimed to determine whether Ad ICmediated DC maturation could be circumvented using these Advector candidates. Results: We found that all Ad vectors tested forming ICs with plasma containing serotype-specific NAbs had the capacity to 1) mature human DCs as monitored by the up-regulation of co-stimulatory molecules and the release of pro-inflammatory cytokines (TNF-a), via the stabilization of Ad capsid at endosomal but not lysosomal pH rendering Ad DNA/TLR9 interactions possible and 2) potentiate Ad-specific CD4 and CD8 T cell responses. Conclusion: In conclusion, despite a conserved DC maturation potential, the low prevalence of serotype-specific NAbs renders rare Ad vectors attractive for vaccine strategies.

POSTERS 214 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.21


Improving the Immunogenicity Against HIV-1 Subtype C Env/Gag-Pol-Nef Proteins Expressed from MVA by the Deletion of the Viral AntiApoptotic Gene F1L
B. Perdiguero1, C. Gmez1, J. Njera1, M. Esteban1
1

Posters
P19.22
Conserved Elements (CE) Vaccine
G.N. Pavlakis1, M. Rolland7, B.K. Felber1, V. Kulkarni1, B. Mothe2, C. Brander2, J. Termini3, G.W. Stone3, S. Le Gall4, J. Yan5, D.B. Weiner5, S. Manocheewa6, J.V. Swain6, E. Lanxon-Cookson6, J.I. Mullins6
1

Centro Nacional de Biotecnologa, Madrid, Spain

Background: Poxviruses are one of the best characterized and more commonly used viral models for vaccine purposes. Among them, the attenuated strain of vaccinia virus MVA provides a high level of exogenous gene expression and safety and immunogenicity against heterologous antigens. Currently, MVAbased recombinants are being used in Phase I clinical trials for HIV/AIDS and a new clinical trial using this strain has recently been completed in Spain. However, efforts for the optimization of this vaccine candidate continue. The aim of present work was the improvement of MVA as vaccine vector against HIV/AIDS by the deletion of the viral anti-apoptotic F1L gene which encodes a mitochondrial-localized inhibitor of caspase-9 and pro-apoptotic members of the Bcl 2-family (Bak and Bax). The deletion of this gene was motivated by previous observations in which dendritic cells that have phagocytosed infected apoptotic cells can present viral antigens to cytotoxic T cells and induce an immune response. Methods: Using MVA-C as parental virus, which expresses from the TK locus gp120 and Gag-Pol-Nef proteins of HIV-1 (subtype C) under the transcriptional control of the synthetic early / late promoter, we have generated the recombinant MVA-C-deltaF1L in which F1L gene has been replaced by rsGFP gene. Results: In this work we have characterized this vector analyzing several parameters both in vitro (viral growth, stability of HIV antigens, induction of apoptosis and secretion of pro-inflammatory cytokines in different murine and human cell lines) and in vivo. Studies on the immunogenicity of the recombinant virus in the mouse model show that F1L gene deletion quantitatively improved the immunogenicity against HIV antigens in a significant manner both during adaptive and memory phases of the immune response. Conclusion: These observations suggest that deletion of the F1L gene could be a valid strategy for the optimization of MVA as vaccine vector.

National Institutes of Health, National Cancer Institute, Frederick, MD, USA; 2Irsi-Caixa-HIVACAT, Barcelona, Spain; 3 University of Miami, Miami, FL, USA; 4Ragon Institute, Boston, MA, USA; 5University of Pennsylvania, Philadelphia, PA, USA; 6University of Washington, Seattle, WA, USA
Background: We are developing an HIV-1 M group vaccine that focuses immune responses on conserved elements (CE) essential to viral function while precluding responses against immunodominant decoys - targets on the virus that can mutate while retaining function. We argue that we will need to target responses using these criteria to achieve high levels of protection. Methods: We compare immune response of DNA vaccines derived from CE to complete gene products in mice, including HLA transgenics, human DC and T cells ex vivo, and macaques. Results: Two toggled variants of CE DNA were made to address ~99% of HIV-1-M variability. Expression of CEgag was maximized in secretion vectors. Proteolytic processing of p24 CE revealed 87% of the known optimal epitopes. Expression by DC elicited strong CD4 and CD8 responses in human T cells ex vivo. Balb/C and Black6 mice develop CEgag-specific CD4, CD8 and Ab. Macaques generated CD8 responses of the effector phenotype. p24 CE are highly immunogenic in HIV-1 infection and include >30 epitopes restricted by >40 HLA. Controllers had higher avidity (p=0.01) and more cross-reactive responses than non-controllers (p=0.01) to the entire p24 and CE regions. Responses of high functional avidity had a superior ability to recognize peptide variants than low avidity responses (p=0.01). Database frequency was too crude a measure to determine the requirement for conservation of a given AA in p24, attributable in part to HLA imprinting over time. Conclusion: p24 CEgag is immunogenic in HIV-1 infection. Immune responses can be focused on these elements by vaccination, to the exclusion of decoy epitopes.

AIDS Vaccine 2011

215

POSTERS

Posters
P19.23

Topic 19: Vaccine Concepts and Design P19.24


Co-Delivery of Mucosal Chemokine Plasmids in a Systemically Administered DNA Vaccine Elicits Systemic and Mucosal Immune Responses in Rhesus Macaques
M. Kutzler1, K. Kraynyak6, A. Sylvester1, N. Cisper6, A. Ginsberg6, J. Yan6, M. Betts6, D. Carnathan6, S. Yuan6, M. Reuter6, N. Kathuria1, N. Hutnick6, A. Khan2, J. Mestecky3, Z. Moldoveanu3, B. Pahar4, M. Hunter4, P. Marx4, D. Montifiore5, D. Weiner6
2

Development of an In Vitro HIV Priming System for Evaluation of Potential Immunogens Prior to Clinical Testing
A. Gervassi1, H. Horton1
1

Seattle Biomedical Research Institute, Seattle, WA, USA

Background: An in vitro priming system has been developed for the evaluation of candidate vaccine inserts side by side with cells from the same seronegative individual. This system will allow the comparison of epitope specificities of induced T cells against different inserts and the ability of these T cells to control in vitro viral replication. Methods: Monocyte-derived dendritic MDDC cells were generated by plastic adherence of PBMC and culture in GM-CSF and IL4. After 24 hours, MDDC were matured by the addition of IL-1, IL-6, TNF- and PGE2 and further incubated for 24 hours. CD45ROneg (nave), CD8pos cells from the same donor were cultured with irradiated, peptide-pulsed MDDC, in media containing IL-21. Every 2-3 days, IL-2, IL-7 and IL-15 was added. Cells were re-stimulated with peptide-pulsed autologous irradiated PBMC and assessed for specificity by ELISpot. Results: This method of generating MDDC (known as fast DC) produces MDDC of similar phenotype as the standard 7-day culture of adherent or CD14pos monocytes as measured by expression of HLA DR and CD11c. Addition of maturation cocktail, significantly increases expression of CD83. These MDDC are efficient at priming HIV-specific CD8+ T cells in vitro. An average of 5 to 30% of wells contained in vitro primed HIV RK-9 and SL-9 peptide-specific T cells after two rounds of stimulation as measured by IFN- ELISpot. Conclusion: These data demonstrate that HIV-specific responses can be primed in vitro from HIV-seronegative donors. Further development of this in vitro priming system will allow us to define characteristics of immunogen inserts that are likely to be important for next generation HIV vaccine design and could be used as a first screen of candidate T cell based immunogens prior to moving forward into more costly human clinical trials.

Drexel University College of Medicine, Philadelphia, PA, USA; Inovio Biomedicals, Blue Bell, PA, USA; 3University of Alabama at Birmingham, Birmingham, AL, USA; 4Tulane National Primate Research Center, Covington, LA, USA; 5Duke University, USA; 6University of Pennsylvania, Philadelphia, PA, USA
1

Background: The induction of mucosal immunity is a crucial goal for HIV vaccines, as the mucosa is the primary site of HIV transmission/viral replication. DNA vaccines which are non-live/non-proliferating have had limited success in this area. Mucosal immune cell homing is in part controlled by a subset of chemokines that include CCL27, CCL28 and CCL25. We hypothesized that mucosal immunity could be generated following chemokine co-delivery with antigens. Methods: Using a rhesus macaque model, we created optimized rhesus CCL27, CCL25, and CCL28 plasmid adjuvants, and coimmunized with optimized/consensus macaque pol and sooty mangabey consensus gag/env antigens. Female macaques were immunized intramuscularly followed by electroporation (IM/EP) with antigenic plasmids plus/minus a single chemokine. Results: Chemokine adjuvants boosted memory responses in an IFN-gamma ELISPOT over antigenic plasmids alone, and coimmunization elicited detectable and long-lived polyfunctional CD8+ T cells responses in the periphery as well as mucosal sites, including the intestine and vagina. Titers of antigen-specific IgA in sera and genital washes of chemokine-vaccinated macaques were observed with the CCL27 and CCL28 adjuvants leading to both neutralization and non-neutralizing B cell phenotypes. A repeated low dose intravaginal challenge with SmmE660 was carried out, and chemokine adjuvants were able to reduce viral set point vs. controls. Importantly, we have characterized the vaccine-specific immune responses to determine the functionality and phenotype of vaccine-induced T and B cells from the mucosal and systemic compartments. Conclusion: The results of this study will be critical to the development of an effective vaccine against HIV. This is the first example of the use of mucosal chemokines to influence a DNA vaccine strategy, suggesting a novel approach for manipulation of vaccine-induced immune responses. This work is supported by funding through the NIH/NIAIDS (F32AI054152 to MA, HIVRADP01AI0739 to DBW).

POSTERS 216 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.25


Efficacy of HIV Vaccine Candidate in Macaques Dependent on the Dose of SIVmac251 Challenge Exposure: Immune Correlates of Vaccine Efficacy
G. Franchini , M. Vaccari , B. Keele , M. Doster , Z. Ma , A. Hryniewicz1, T. Morgan1, Y. Guan4, G. Ferrari5, D. Montefiori5, D. Venzon6, C. Fenizia1, J.D. Lifson7, C. Miller3, J. Tartaglia8
1 1 2 1 3

Posters
P19.26
Biologically Active Fusion Proteins of CD154 and SIVgp41 as Novel Vaccine Components
L.M. Smith2, L.M. Parodi2, V.L. Hodara2, G. Pavlakis1, B. Felber1, L.D. Giavedoni2
1 2

Center for Cancer Research/NCI/NIH, Frederick, MD, USA; Texas Biomedical Research Institute, San Antonio, TX, USA

National Cancer Institute/ National Institute of Health, Bethesda, MD, USA; 2AIDS and Cancer Virus Program, NCIFrederick, Frederick, MD, USA; 3California National Primate Research Center, USA; 4Human Monoclonal Antibody Core Laboratory, University of Maryland, USA; 5Duke University Medical Center, USA; 6Biostatistics and Data Management Section/ NCI/NIH, USA; 7Human Retrovirus Section, NCIFrederick, MD, USA; 8sanofi pasteur, Inc, Swiftwater, PA, USA
1

Background: Mucosal transmission of HIV involves a limited number of viral variants, suggesting there may be a window of opportunity for an HIV vaccine. Thus, SIVmac251 infection of macaques, a widely used model in the assessment of relative efficacy of vaccine candidates for HIV, should recapitulate transmission of HIV to humans. Methods: We investigated the impact of the mucosal SIVmac251 challenge dose on vaccine efficacy in twenty macaques that were immunized with a combination of DNA, ALVAC-SIV, and the gp120 envelope protein vaccines. The animals were exposed intrarectally to SIVmac251 either as a single dose of 6100 TCID50, or a weekly dose of 470 TCID50 of the same virus stock until all controls became infected. Results: Vaccination did not protect from infection with a challenge dose of 6100 TCID50, but did result in a lower plasma virus in vaccinated versus control animals over the first three weeks of infection (p=0.010;). In contrast, two exposures of the remaining twenty-four macaques to 470 TCID50 of SIVmac251 resulted in infection of all control animals but only nine of the twelve vaccinated macaques (25% efficacy). The remaining vaccinated animals that became infected had a significant and durable reduction in plasma virus level, were protected from CD4+T-cell loss, and had a significant reduction in the number of transmitted virus variants (an average of 1). Vaccine-induced immune responses that correlated with protection from disease were: pre-challenge Gag and Envelope specific ELISpot responses, Gag lymphoproliferative responses, and Gag specific CD8+CD107+ T-cells producing TNF-. A significant inverse correlation was found between virus levels and gp120 antibody avidity and Antibody Dependent Cellular Cytotoxicity. Conclusion: Accurate modeling of SIV transmission in macaques is essential in the evaluation of vaccine efficacy. Durable vaccine protection from high levels of viral replication requires both Band T-cell responses.

Background: HIV gp41 includes conserved residues critical for the function of the viral spike, which suggests that these regions might be important targets for neutralizing antibodies; however, the poor immunogenicity of this region may be due to its lack of exposure on native virus. CD154 (CD40L) is a trimeric glycoprotein found on activated CD4 T-cells that binds to CD40 on APCs and leads to B-cell activation and differentiation to plasma cells. We aimed at improving immunogenicity of multigenic prime/boost vaccination approaches by designing a fusion protein of the CD40-binding domain of CD154 and gp41. This protein would assemble into trimers with the potential for inducing neutralizing antibodies to an exposed gp41 and stimulating activity on APC and B-cells Methods: CD154 and SIV gp41 are trimeric glycoproteins with opposing polarities. A flexible (FL), helical (HL), and random linker (NL) were used to join both protein domains. Stable cell lines were prepared that expressed each one of these fusion proteins, along with SIV Gag for production of VLPs that contained the fusion proteins. We also generated fusion proteins that contained SIV gp41 with shorted cytoplasmic tails (CT) and recombinant Vaccinia viruses (VV) that expressed these fusion proteins. Results: Both the FL and HL allowed proper protein folding and biological activity. Surprisingly, VLPs containing the short gp41 CT incorporated lower amount of fusion protein than the ones with full length gp41. Purified VLPs were able to induce cytokine expression from macaque PBMCs. VV expressing CD154/gp41 proteins also induced expression of cytokines associated with CD40 ligation from macaque PBMC. Conclusion: Fusion proteins of CD154 and gp41 were made that assembled into trimers, incorporated into VLPs and activated immune cells. The ability of these fusion proteins to induce neutralizing antibodies will be tested in nonhuman primates.

AIDS Vaccine 2011

217

POSTERS

Posters
P19.27

Topic 19: Vaccine Concepts and Design P19.28


Carbohydrate-Based HIV Vaccine, from Concept to Mannose-Specific Virion-Binding Antibodies
Y. Geng1, H. Fu1, B. Liu1, H. Zhang1, R.J. Luallen1, R.W. Doms2
3 1

Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) Is a Correlate of Protection by LiveAttenuated SIV


M.D. Alpert , J.D. Harvey , W.A. Lauer , R.K. Reeves , M. Piatak , A. Carville4, K.G. Mansfield4, J.D. Lifson3, W. Li5, R.C. Desrosiers1, R.P. Johnson2, D.T. Evans1
1 1 1 2 1 New England Primate Research Center, Harvard Medical School, Southborough, MA, USA; 2Immunology Division, NEPRC, Harvard Medical School, Southborough, MA, USA; 3 SAIC Frederick, National Cancer Institute at Frederick, Frederick, MD, USA; 4Department of Pathology, NEPRC, Harvard University, Southborough, MA, USA; 5University of Massachusetts, Worcester, MA, USA

ProSci Inc., Poway, CA, USA; 2University of Pennsylvania, Philadelphia, PA, USA

Background: Several lines of evidence indicate that the glycan shield may be a rewarding HIV-1 vaccine target. Over half-dozen constructs targeting the glycan shield have been developed in attempts to induce 2G12-like neutralizing antibodies. Methods: A triple mutant (TM) strain of Saccharomyces cerevisiae was engineered by deleting three genes in the N-glycan pathway. Glycan profiling was analyzed by MALDI-TOF, and 2G12-cross reactive proteins were detected by Western blots and identified by nano-LC-MS/MS. Rabbit antisera were raised with TM yeast cells or single 2G12-reactive yeast glycoprotein, and tested by ELISA, Western blots and glycan microarray. HIV-1 pseudoviruses were generated in 293T cells, and neutralization assay was performed using U87.CD4.CCR5 or TZM-bl cells. The binding of the antisera to pseudovirions was tested by a viral capture assay. Results: The TM yeast cell expresses almost exclusively Man8GlcNAc2 N-linked glycans on glycoproteins. Five endogenous yeast glycoproteins that efficiently bound to 2G12 were identified. Unlike human glycoproteins, these 2G12reactive yeast glycoproteins contain a large number and high density of N-linked glycans, similar to gp120. Immunization of rabbits with whole TM yeast or single 2G12-reactive yeast glycoprotein elicited antibodies that specifically bound to the clusters of terminal Man1,2-Man 1,2-Man oligosaccharides. More importantly, the mannose-specific antibodies were able to bind to a broad range of monomeric gp120 from different HIV subtypes and SIV. Notably, the antibodies could also bind to the pseudovirions and efficiently neutralize a genetically diverse panel of HIV-1 pseudoviruses when the viruses were produced in the presence of a mannosidase inhibitor kifunensine to force retention of high-mannose Man9GlcNAc2 N-linked glycans. Conclusion: Mannose-specific HIV-1 Env cross-reactive antibodies can be elicited with 2G12-reactive yeast glycoproteins or whole TM yeast. This genetically manipulated yeast strain is a powerful, feasible, and cost-effective means to produce glycoantigens to recapitulate the 2G12 epitope for the development of a carbohydrate-based HIV-1 vaccine.

Background: Live-attenuated strains of simian immunodeficiency virus (SIV) afford apparent sterilizing immunity against pathogenic SIV challenge with the greatest reliability achieved thus far in non-human primate models. Identification of the mechanisms responsible may provide insights important to the design of a safe and effective HIV-1 vaccine. Methods: We measured the capacity of antibodies elicited by nefdeleted strains of live-attenuated SIV to direct ADCC against SIVinfected cells. Protection afforded by SIV nef was studied in two SIVmac251 challenge experiments. One was designed to investigate temporal associations with protection, and the second addressed the effect of sequence similarity in Env with the vaccine strain on the extent of protection. Results: Env-specific ADCC titers against SIV-infected cells emerged early and increased over time, whereas neutralization was largely undetectable. The development of these antibodies depended upon vaccine persistence and was proportional to the extent of SIV nef replication. ADCC titers were higher against SIV strains matched in Env with the vaccine strain than against Env-mismatched strains. In both SIVmac251 challenge experiments, the macaques vaccinated with SIV nef that remained uninfected had significantly higher ADCC titers against SIVmac251-infected cells than those that became infected. Higher ADCC titers were also associated with lower peak viral loads in the infected animals. Conclusion: These results identify ADCC as a correlate of protection by live-attenuated SIV, and suggest ADCC may be a useful activity to elicit in designing vaccines to prevent HIV-1 infection.

POSTERS 218 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.29


Focusing the Antibody Response to the CD4Binding Site with Designer Immunogens
F.K. Ahmed1, B. Clark1, D.R. Burton2, R. Pantophlet1 Simon Fraser University, Burnaby, Canada; 2Dept of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
1

Posters
P19.30 LB
Preexisting Immunity to Multiple Adenovirus Serotypes Is Not Associated with Increased HIV-1 Acquisition in Three HIV-1 Vaccine Efficacy Trials
K.E. Stephenson1, J. Hural2, S.P. Buchbinder3, F. Sinangil4, D.H. Barouch1
1

Background: The HIV envelope glycoproteins gp120 and gp41 are the primary targets for neutralizing antibodies (NAbs), and thus of considerable interest for vaccine design. Of particular interest is the CD4-binding site (CD4bs) on gp120 which is targeted by bNAbs such as b12, VRC01/02 and HJ16. Although the elicitation of CD4bs-specific NAbs has been reported upon experimental immunization, such antibodies are typically of low titre and transient. Approaches to improve the potency and durability of CD4bs-specific responses are needed. Methods: In earlier work, a panel of so-called hyperglycosylated gp120s were generated. These mutant gp120s are engineered to expose the b12-binding site while occluding the epitopes of numerous non-NAbs. Here, we present data on the effects of two adjuvants, monophosphoryl lipid A (MPL) and QuilA, on improving CD4bs-specific responses when mixed with one of the hyperglycosylated mutants, N2mCHO(Q105N), in comparison to wild-type gp120 (gp120wt). To dissect CD4bsdirected specificities, a truncated gp120 outer domain construct (XOD6) and a resurfaced core protein (RSC3) were utilized. RSC3 specifically presents the neutralizing face of the CD4bs but largely eliminates other antigenic faces through non-HIV-1 residue substitution. Results: gp120wt sera from MPL and QuilA animals bound strongly to the homologous antigen but poorly to Q105N, indicating that Q105N indeed limits exposure of undesired epitopes. Q105N sera from MPL animals also bound the homologous antigen preferentially, but this preference was less pronounced with sera from QuilA animals. Sera from all animal groups bound XOD6 at similar levels. However, the QuilA groups showed stronger binding to RSC3 whereby the combination of Q105N with QuilA elicited modestly greater CD4bs-directed responses than gp120wt. Conclusion: Overall, this study demonstrates the effect of adjuvants on improving both titre and quality of CD4bs-directed responses. Studies are ongoing to determine the fraction and binding specificity of CD4bs-specific antibodies in the sera and analyze their neutralization activity.

Division of Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; 2Fred Hutchinson Cancer Research Center, Seattle, WA, USA; 3San Francisco Department of Public Health, San Francisco, CA, USA; 4Global Solutions for Infectious Diseases, South San Francisco, CA, USA

Background: The Step study of the MRKAd5 HIV-1 gag/pol/nef vaccine raised the possibility of enhanced HIV-1 acquisition risk in vaccinees who were both Ad5 seropositive and uncircumcised. Here we examined whether preexisting immunity to multiple adenovirus serotypes was intrinsically associated with enhanced HIV-1 acquisition in the VAX003, VAX004, and Step studies. Methods: We performed case-controlled studies to assess the association between baseline neutralizing antibodies (NAbs) to Ad serotypes 1, 2, 5, 6, 26, 35, and 48 and subsequent HIV-1 infection among 1,570 adults enrolled in the VAX003 and VAX004 trials of the HIV-1 rgp120 vaccine and the Step study of the MRKAd5 vaccine. Cases represent all subjects who became HIV-1-infected. Controls were matched for demographic variables, and, for the Step study, also Ad5 serostatus and circumcision status. Results: NAb titers to Ad serotypes 1, 2, 5, 6, 26, 35, and 48 were determined for 669 HIV-1-infected cases and 901 matched HIV-1-uninfected controls. Ad seroprevalence ranged from rare (Ad35, 9%) to common (Ad2, 87%) and proved comparable between cases and controls (< 5% difference) for all serotypes. There was no association between baseline NAbs and HIV-1 acquisition for any of the seven serotypes tested (P > 0.05 for all comparisons). Odds ratios ranged from 0.8 to 1.2 and 95% CI crossed 1.0 for all serotypes. Sub-analyses by trial, vaccine/ placebo, and circumcision status also found no association (P > 0.07 for all comparisons, adjusted for multiplicity.) Conclusion: Ad5 seropositivity was not associated with increased risk of HIV-1 acquisition in the VAX003 and VAX004 studies. Moreover, seropositivity to six other adenovirus serotypes was not associated with increased HIV-1 acquisition in the VAX003, VAX004, and Step studies. These data demonstrate that Ad seropositivity is not intrinsically a marker for increased risk of HIV-1 acquisition, including following vaccination with rgp120 or an Ad vector from a different serotype.

AIDS Vaccine 2011

219

POSTERS

Posters
P19.31 LB

Topic 19: Vaccine Concepts and Design P19.32 LB


HIV-1 JR-FL Env Trimers Expressed by DNA Electroporation Alone or Boosted With Soluble Trimers Elicit Neutralizing Antibodies
B. Chakbrabarti2, K. McKee1, J. Mascola1, R.T. Wyatt2
1

Pool Approach and Variability Analysis in Isolation and Characterization of HIV-1 Subtype A1 Strain from Russia
A. Galkin1, G. Kuznecovs2, A. Bychenko3, N. Polyakov4, E. Filinova3
2

Advanced Biomedical Research Laboratory, Moscow, USA; Advanced Biomedical Research Laboratory, Physics Method of Analysis D, Moscow, Russian Federation; 3Advanced Biomedical Research Laboratory, Cellular Biology/Virology Dep, Moscow, Russian Federation; 4Bioorganic Chemistry Institute Named by Shemyakin and Ovchinnikov, Moscow, Russian Federation
1

Vaccine Research Center, Bethesda, MD, USA; 2IAVI Neutralizing Antibody Center at The Scripps Research Institute, La Jolla, CA, USA

Background: Library approach for native patients isolates/lab. strain HIV obtaining, genetic and MS-MS variability analysis, in vitro-in vivo multiplication dynamics study were used for tailoring gp120-gp160 immunogenic composition Methods: HIV strain was isolated from patients cohort 4 weeks cultivation with donors PBMC in presence of FGA, IL-2 and adapted to MT-4 or U937 background 40-90 passages cultivation. Full-length genome was amplified by RT-two-round PCR and sequenced. PEG-precipitation, ultracentrifugation 160000g 45min. sucrose 1,16-1,18 g/sm3 gradient, HIV-1-gp160-specific phage-presented libraries or native human polyclonal antibodies, SDS-PAGE bands, tryptic cleavage, consequently, were used for viral proteins concentration. One-demensional LC-MS-MS Esquire-6000Plus and PEAKS software peptides identification were performed for envelop mapping and primers arrangement. Pools of PCR products with 6His were tailored into pLEXSY_Ineo2 vectors and secretory tet-inducible L.tarentolae T7-TR host (400mV, BTX-830,). Env proteins were extracted with ultrafiltration 30 kDa Pelicon membrane and LC 20ml HisTrap or immobilized Ni-NTA (Superose12, GE). Results: HIV-1 subtype A laboratory strain designated PokA-79 was isolated from the pool of 24 HIV-1 infected Russian patients PBMC and MT4, U937 cultures. Single HIV-infected PBMC cultivation did not produce isolates with positive multiplication dynamics in vitro. Entire genome sequence was obtained (NCBI acc. # FJ864679), it belonged and clustered with other A1 subtype Former Soviet Union (FSU) sequences. PokA-79 gag, pol and env sequences were distinguished significantly from corresponding initial primary 24 patients isolates. Some of PokA79 genes contain insertions and frame shifts, gp120 V3 loop has rare GHT insertion similar to several HIV-1 A1 sequences from central Africa. In vitro dynamics with titres 1011-1012 c/ml was less severe but similar to U455 A1 strain, in vivo challenging on MT4-U937 background provided 104-106 c/ml in SCID mice bloodstream. Primers were assembled comprising V1-V5 loops beginning variability forward and C5 end reverse. Conclusion: Rec. gp120-gp160 immunizations provided HIV envspecific bloodstream immune response stable for 2 weeks.

Background: All known HIV-1 gp120-directed broadly neutralizing antibodies efficiently recognize fully cleaved JR-FL spikes, however, the non-neutralizing gp120-directed antibodies cannot. Therefore, as an immunogen, cleaved, functional spikes may selectively present neutralizing epitopes to B cells, perhaps more efficiently eliciting neutralizing antibodies. However, attempts to make soluble versions of Env that fully mimic the viral spike are yet successful. Methods: To present functional, cleaved spikes to the immune system, we inoculated both non-human primates (NHPs) and rabbits with JR-FL Env-expressing plasmid DNA by electroporation. In vitro, this plasmid encodes for fully cleaved, cell surface, trimeric JR-FL Env, but only when expressed from a mini-LTR in a non-codon optimized state. DNA priming was followed by boosts with soluble JR-FL gp140 trimers in adjuvant. In vivo, the LTR-driven, non-codon-optimized Env plasmid DNA, likely requires tat co-transfection in trans, and rev expression in cis, for efficient Env expression. A control codon-optimized and CMV-driven Env plasmid was used; however, in vitro this plasmid expresses inefficiently cleaved JR-FL cell-surface trimers. Results: We report that the DNA priming elicited reasonable ELISA binding titers in both NHPs and, somewhat surprisingly, also in rabbits. The elicitation of neutralizing antibodies in the NHPs was exceedingly high against Tier 1 isolates following protein boosting. In rabbits, neutralization from the non-codonoptimized DNA was dependent upon co-transfection with the tat expressor plasmid, but not so for the codon-optimized construct. Reasonable Tier 1 neutralizing antibodies were elicited in rabbits after 3 DNAs, with modest binding titers, and weak, sporadic neutralization of Tier 2 isolates. The neutralizing activity generally increased following protein boosting. Conclusion: We conclude that DNA priming by electroporation is an interesting means to present functional, cleaved HIV-1 Env spikes to the B cell immune system to prime or initiate trimerelicited neutralizing antibodies in both rabbits and, importantly, primates.

POSTERS 220 AIDS Vaccine 2011

Topic 19: Vaccine Concepts and Design P19.33 LB


Evaluations of Subtype C HIV-1 Envelope Glycoproteins as Immunogens for Vaccine Development
A.K. Dey1, C. Zambonelli1, A. Nandi1, S. Stephenson1, K. Sirokman1, S. Hilt1, D. Martinez-Guzman1, J. Cisto1, P. Ramesh1, M. Wininger1, Y. Sun1, H. Legg1, K. Hartog1, R. Pei-Jen Lai2, J. Heeney3, D. Montefiori3, A. Carfi1, S.W. Barnett1
1

Posters

Novartis Vaccines and Diagnostics, Cambridge, MA, USA; 2Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom (Great Britain); 3 Department of Surgery, Duke University Medical Center, Durham, NC, USA
Background: A successful vaccine against HIV-1 will likely require both a cellular and a neutralizing antibody (nAb) component. Despite substantial efforts to produce a potent vaccine immunogen capable of inducing broadly nAbs, little success has been achieved. The inability to generate an appropriate antigen has been hindered by enormous antigenic variability and immune-evasion mechanisms exhibited by the viral envelope glycoprotein (Env). The screening of new immunogens and their through in vitro and in vivo evaluations provides rationale for immungon selection for future pre-clinical and clinical studies. Methods: Besides rationale antigen design, our approach has been to screen multiple recombinant Envs from acute subtype C HIV-1 isolates for identifying superior antigen that generates better Env specific neutralizing response in small animals. In conjunction, we have also evaluated multivalent and sequential Env vaccinations with novel formulation strategies to elicit improved antibody response. Results: We observe heterogenous expression of gp120s from acute subtype C HIV-1 isolates, predominantly as mixtures of monomer and dimers. The dimers, which are aberrantly disufidelinked proteins, constitute 15-70% of stable cell-line expressed gp120s. Although in vitro, both gp120 forms differ in ligandbinding abilities, in vivo they are similarly immunogenic. Considering >70% monomer and >5mcg/ml expresssion and other criteria, two gp120 proteins were selected for future production. Conclusion: The process of screening, in vitro characterization, generation of stable cell lines and immunogenic evaluations of the proteins in various small animals have led to the selection of top two HIV-1 subtype C gp120s for production to support Pox prime-Protein boost Phase 2b clinical trial in Southern Africa.

AIDS Vaccine 2011

221

POSTERS

Posters
P20.01

Topic 20: Viral Diversity P20.02


HIV Integrase Polymorphisms in Treatment-Nave and Treatment-Experienced HIV-Infected Patients in Thailand Where HIV Subtype A/E Predominates
A. Phuphuakrat1
1 Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand

Non-Random Distribution of Cryptic Repeating Triplets of Purines and Pyrimidines (RNY)n and Recombination in gp120 of HIV-1
E. De Crignis1, S. Guglietta1, B.T. Foley2, M. Negroni3, A.F. Di Narzo4, V. Waelti Da Costa5, M. Cavassini5, P. Bart1, G. Pantaleo1, C. Graziosi1
1

Lab. of AIDS Immunopathogenesis, Div. of Immunology and Allergy,CHUV, Lausanne, Switzerland; 2Theoretical Biology Biophysics Group-Los Alamos National Laboratories, Los Alamos, NM, USA; 3Architecture et Ractivit de lARN,Universit de Strasbourg,CNRS,IBMC, Strasbourg, France; 4 Swiss Institute for Bioinformatics - Bioinformatics Core Facility, Lausanne, Switzerland; 5Division of Infectious Diseases, Department of Medicine, CHUV, Lausanne, Switzerland
Background: The tendency of HIV-1 to undergo sequence variation, particularly in gp120, is one of the major impediments to the development of a successful preventive HIV vaccine. We have previously shown that region V4 of gp120 is characterized by high levels of intra-host length polymorphism due to insertions/ deletions (indels) of multiple of three base pairs. In this study we have characterized intra-host variation in all regions of gp120. Methods: Cloning and sequencing of a fragment of env spanning C1-C5 derived from plasma RNA of seven patients with early infection and nave to therapy. Recombination analysis was performed using Splitstree and the RDP3 package. Statistical data were obtained by a randomization test using an in-house R script in the R environment. Results: Major insertions/deletions (indels) were found in gp120 in V1, V2, V4, and V5, and to a lesser extent in C3, altering the glycan profile of the regions in which they occurred. Inserted/ deleted fragment consisted of duplications and stretches of repeats of the trinucleotides RRY and RNY (R=purine, Y=pyrimidine, N = any nucleotide), suggesting a possible involvement of these trinucleotides in the genesis of indels. Statistical analysis showed a non-random distribution of strings of (RNY)n, with longer strings significantly more frequent in variable than in constant regions. Recombination was detected in all patients. Due to recombination events, polymorphic constant and variable regions of gp120 were found to rearrange in individual clones of the quasispecies, so that each region of gp120 appeared to evolve independently Conclusion: Cryptic repeats of purines and pyrimidines appear to play an important role in the generation of intra-host genetic diversity of gp120. Mutations due to length polymorphism spread through the quasispecies via recombination. This study provides evidence of the dramatic molecular complexity that characterizes the intra-patient variability of gp120 in vivo.

Background: Integrase inhibitor (INI) is a novel antiretroviral drug recommended for both treatment-nave and treatmentexperienced HIV-1-infected patients. Limited data are available on INI resistance in Thailand, where HIV-1 subtype A/E predominates. We aimed to investigate INI resistance-associated mutations (RAMs) among treatment-nave patients and patients who experienced treatment failure with nonnucleoside reverse transcriptase inhibitor (NNRTI)-based or protease inhibitor (PI)based antiretroviral therapy (ART) in Thailand. Methods: One hundred and eight plasma samples of 58 treatment-nave and 50 treatment-experienced HIV-1-infected individuals were collected. HIV-1 integrase coding region was sequenced. Polymorphisms were compared between subtype A/E and B circulated in Thailand and between treatment-nave and treatment-experienced groups. Resulting amino acids were interpreted for drug resistance according to Stanford algorithms. Results: Ninety-seven samples were HIV-1 subtype A/E; 10 were subtype B and one was subtype C. Age, gender, and CD4 cell counts were similar between treatment-nave and treatment-experienced groups. Major INI-RAM was not found in this study, but some minor INI-RAMs, such asV54I, L68I, L74M, T97A and S230N, were found. Comparing INI-RAMs between subtype A/E and B, prevalence of V51I and V72I was higher in subtype B than subtype A/E, while V201I was found in all sequences of subtype A/E. In subtype A/E, integrase polymorphisms were not different between treatment-nave and treatment-experienced groups. However, number of amino acid substitutions were significantly higher in treatment-experienced group (P = 0.009). One NNRTI-based ART treated patient was found to have potential low-level INI-RAMs. Conclusion: INI-RAMs are rare in both treatment-nave and treatment-experienced patients in Thailand. This suggested that INI should be active in patients who are nave to INI in Thailand.

POSTERS 222 AIDS Vaccine 2011

Topic 20: Viral Diversity P20.03


Comparable Antigenicity Among Subtype B, Subtype F1 and Group M Nef Consensus Peptides in Brazilians HIV-1-Infected Subjects
F. Crtes1, G. Bello1, C. Vorsatz 2, J. Pilotto1, B. Grinsztejn2, V.G. Veloso2, S.W. Cardoso2, M.G. Morgado1
1 2

Posters
P20.04
Characterization of HIV-1 Subtype Distribution Among Thai MSM Using MHAbce, a High Throughput Approach for Molecular Epidemiology Studies
W. Leelawiwat1, W. Rutvisuttinunt2, J. McNicholl3, M. Arroyo2, B. Raengsakulrach1, F. Mueanpai1, O. Kongpechsatit1, V. Assawadarachai2, M. de Souza2, S. Chaikummao1, W. Chonwattana1, J. Tongtoyai1, A. Sangiamkittikul1, M. Curlin3, F. van Griensven3 Thailand Ministry of Public Health-U.S. CDC Collaboration, Nonthaburi, Thailand; 2Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand; 3Centers for Disease Control and Prevention, Atlanta, GA, USA
1

Oswaldo Cruz Institute/FIOCRUZ, Rio de Janeiro, Brazil; Evandro Chagas Clinical Research Institute/FIOCRUZ, Rio de Janeiro, Brazil

Background: The use of centralized viral sequences has been proposed to reduce genetic distance between vaccines antigens and circulating viruses. In Brazil the B subtype is predominant followed by subtype F1 and B/F1 recombinants. The aim of the present study is to compare T cell responses to Nef HIV-1 peptides based on consensus and viral isolate sequences from B and F1 subtypes and group M consensus. Methods: Fifty clade B and 36 clade F1 Brazilian sequences were aligned with CLUSTALW, and the corresponding subtype B and F1 consensus sequences were retrieved using DAMBE v.5.0 program. Sequences from those subtypes B and F viral isolates presenting the minor genetic distance to the set of sequences were also selected. The group M consensus was obtained from Los Alamos database. Nef-specific T cell responses were evaluated with IFN- ELISpot assay to three overlaid regions (R). PBMCs from 18 and 9 subjects infected with HIV-1 subtypes B and F1, respectively, were analyzed. Results: A high cross-reactivity among subtype B, subtype F1 and group M Nef consensus peptides was detected. More than 70% of subjects had a positive response against all consensus peptides to at least one region analyzed. The evaluation of frequency and magnitude of responses also demonstrated that peptides sets based on consensus and isolates were similarly powerful in detecting intra-clade T cell responses. However, the frequency of inter-clade T cell responses to isolates was smaller than the corresponding responses to consensus in R2 (subtype B and subtype F1-infected subjects) and R3 (subtype B-infected subjects). Conclusion: Our results suggest that Nef peptide sequences based on HIV-1 clade B, F1 or group M consensus are more appropriate for the design of immunogens than peptide sequences based on single viral isolates from the Brazilian epidemic.

Background: Understanding the local molecular epidemiology of HIV-1 subtypes may be important for the development of HIV vaccines. Here, we describe the HIV-1 subtype distribution in a cohort of MSM in Bangkok, followed from April 2006September 2009. Typing was performed using MHAbce, an RTPCR-based assay using subtype-specific probes targeting 8 gene regions. Methods: Blood samples from 278 seroprevalent and 99 seroincident cases were analyzed. Among seroincident cases, MHA was performed on the first available blood-specimen after seroconversion. Samples were non-typeable when hybridization occurred at <4 loci. In apparent cases of dual infection, cloning and targeted genomic sequencing was performed to verify MHA results. Among seroincident cases, non-typeable and apparent recombinant cases were verified by direct sequencing of MHA amplification products. Results: In 16 apparent dual infections, sequence verification confirmed the presence of two subtypes in 5, single infection in 2, and highly diverse sequences in 9. Sequencing confirmed a recombinant genome in 8/10 seroincident recombinants. ln 6 non-typeable cases, MHA and sequencing results were entirely concordant. After confirmation, the overall distribution of HIV1 subtypes was: CRF01_AE, 81.6%; CRF01_AE/B recombinants, 11.0%; B, 4.9%; dual CRF01_AE and B, 1.5%; B/C/CRF01_AE recombinants, 0.6%; and B/C recombinants, 0.3%. The subtype distribution did not vary significantly by year or by serostatus (incident vs. prevalent). Conclusion: In this cohort of HIV-1 infected Thai MSM, CRF01_ AE was the most common and B was least common subtype. All remaining infections were caused by complex recombinant forms. The predominance of CRF01_AE is similar to that currently observed in HIV-1 infected Thai heterosexuals and IDU. The presence of complex recombinant forms is probably due to rapid sexual partner turnover among MSM. While the predominance of CRF01_AE may seem to simplify the design of an HIV-1 vaccine for Thailand, the presence of multiple complex recombinant forms may pose new challenges.

AIDS Vaccine 2011

223

POSTERS

Posters
P20.05

Topic 20: Viral Diversity P20.06 LB


Genetic Analysis of the Viral Envelope (V3-V5 region) in HIV-1 Infected Indian Children
P. SS1, R. Kalra1, R. Lodha2, S. Kabra2, K. Luthra1
1 2

Evolution of Nigerias Two Predominant HIV-1 Subtypes


H.O. Ajoge1, M.L. Gordon2, T. de Oliveira3, S. Ibrahim4, O.S. Shittu5, S.O. Olonitola1, A.A. Ahmad1, T. Ndungu2 Department of Microbiology, Ahmadu Bello University, Zaria, Nigeria; 2HIV Pathogenesis Programme, DDMRI, University of KwaZulu-Natal, Durban, South Africa; 3Africa Centre for Health and Pop. Studies, University of KwaZuluNatal, Durban, South Africa; 4Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; 5Department of Gyneacology and Obstetrics, ABUTH, Ahmadu Bello University, Zaria, Nigeria
1

All India Institute of Medical Sciences, New Delhi, India; Pediatrics, AIIMS, New Delhi, India

Background: HIV-1 infection in India is predominantly due to clade C viruses. Our objective was to assess the variability in the V3-V5 region of viral envelope from HIV-1 infected Indian children. Methods: Thirteen HIV-1 infected children were recruited after getting written informed consent from the parents/guardians. The study was approved by the institute ethics committee. V3-V5 region of the HIV-1 envelope was amplified from the genomic DNA by nested PCR, purified and then cloned into T/A cloning vector. Random positive clones were sent for sequencing commercially (Macrogen Inc, Korea). HIV-1 subtyping was done using REGA HIV-1 subtyping tool available in Stanford HIV database. Coreceptor usage was predicted using Web PSSM. Variability in the V3, V4 and V5 region was analyzed by aligning the sequences in Clustal X. Results: All of the 13 children were infected with HIV-1 subtype C. V3 region was found to be highly conserved with GPGQ motif present typical of subtype C viruses. Predicted coreceptor usage was CCR5 for all the 13 viruses except for one sample (AIIMS_307) which showed CXCR4 usage. This patients V3 sequence was 37 amino acids long with insertion of 2 amino acids and a GRGQ motif. A salient finding of this study is the complete deletion of the envelope V4 region of the virus infecting one patient (AIIMS_363). The number of potential N-linked glycosylation sites in the V3-V5 region of this virus was only 8 while in the rest of the patients they were in the range of 12-17. GenBank accession numbers: HQ845212, HQ845213, HQ845224, HQ854231, HQ854232 and JN176191-JN176198. Conclusion: We found CXCR4 usage in one and a V4 deleted envelope in another virus from the subtype C infected children from north India. Genetic analysis should be done in all infected patients to elucidate the molecular epidemiology of HIV-1 infection in this region.

Background: HIV-1 CRF02_AG and subtype G (HIV-1G) account for most HIV infections in Nigeria, but their evolutionary trends remains obscure. Methods: CRF02_AG gag and env data sets consisting of 28 and 33 sequences respectively, dating from 1994 to 2007; and HIV1G gag and env data sets consisting of 23 and 50 sequences respectively, dating from 1992 to 2007 was utilized. The data sets consisted of the only available dated Nigerian gag and env sequences at time of study. To determine the origin of Nigerian CRF02_AG and HIV-1G epidemics, Maximum likelihood phylogenetic analysis (MLPA) was carried out on the data sets and reference sequences from different geographic origins. Bayesian analysis was carried out on data sets with well-supported Nigerian clade(s) in the Maximum likelihood trees (MLT). Results: MLPA suggested multiple independent entries of both subtypes into Nigeria. HIV-1G gag and env trees had one and two well-supported clades (bootstrap >70%), respectively; while CRF02_AG trees had none. The best fit models, relaxed Bayesian skyline plot (BSP) and relaxed constant population size models, estimated Nigerian HIV-1Gs most recent common ancestor to 1973.2 [Highest probability densities(HPDs) 1953.4-1984.9] and 1957.3 [HPDs 1867.4-1982.9], respectively, based on gag data set; and to 1971.6 [1960.1-1980.3] and 1969.5 [HPDs 1955.6-1979.3], respectively, based on env data set. The BSP traced the origin of the Nigerian transmission clades in the MLT to 1974 (gag) and 1981 (env). Based on BSP, HIV-1G epidemic involved a three phase growth (pre-1985 constant population, an exponential growth from 1985 to early-1990s, and a slower post early-1990s growth). Conclusion: This study (for the first time) observed that both subtypes made multiple independent entries into Nigeria and that HIV-1G has been in Nigeria since 1980 or earlier. More studies involving archive and recent strains and full-length genome sequences are necessary to fortify our inferences.

POSTERS 224 AIDS Vaccine 2011

Topic 20: Viral Diversity P20.07 LB


Significant Contribution of Subtype G to HIV-1 Genetic Complexity in Nigeria
R.A. Heipertz, Jr.1, O.R. Ayemoba2, E. Sanders-Buell1, J. Kuroiwa1, T. Cervenka1, S. Peel1, J. Malia3, M. Maisaka4, D. Singer5, M. Robb1, J. Kim1, N. Michael1, O.S. Njoku6, S. Tovanabutra1 US Military HIV Research Program, Rockville, MD, USA; Emergency Plan Implementation Committee, Nigerian Ministry of Defense, Abuja, Nigeria; 3US Military HIV Research Program, United States Public Health Service, Rockville, MD, USA; 445 Nigerian Air Force Hospital, Makurdi, Nigeria; 5DoD HIV Program (Nigeria), Abuja, Nigeria; 6US Military HIV Research Program (HJF-MRI), Abuja, Nigeria
1 2

Posters
P20.08 LB
Generating Viruses Matching RV144 Vaccine Components to Explore Efficacy
A. Chenine1, E. Sanders-Buell1, A. Bradfield1, E. Lei1, T. Towle1, W. Murtaugh1, R. Mclinden1, D. Pillis1, M. Wesberry1, V. Polonis1, D. Montefiori2, N. Michael1, J. Kim1, S. Tovanabutra1
1 Henry Jackson Foundation, Rockville, MD, USA; 2Duke Uuniversity, Durham, NC, USA

Background: Global vaccine concepts either include the subtypes A, B, C and CRF01_AE for insert design or mosaic inserts derived from published databases with low West African subtype representation. The conventional view that West African HIV variants are largely CRF02_AG would justify the inclusion of subtype A sequences in a global design. We challenge this justification by presenting the significant contribution of subtype G in the HIV-1 epidemic in four Nigerian cities using a multiregion hybridization assay (MHA) that can identify and differentiate between subtype G and CRF02_AG. Methods: Viral RNA was extracted from plasma of 71 volunteers for a cohort for prevalence, risk factor, and subtype study conducted in Nigeria. Twelve were from Kaduna, 18 from Abuja, 30 from Makurdi, and 11 from Enugu and all were subjected to the G/CRF02_AG MHA, a high throughput assay. This MHA was designed to contain 7 regions: 3 (pol [RT], pol [INT], tat) identify both subtype G and CRF02_AG, and 4 (gag, pol [RT2], vpr, env [gp120]) differentiate between the two subtypes. For differentiating regions, probes are designed to be subtype specific. Results: Overall, subtype G represents 23% while CRF02_AG contributes 41% and their recombinants represent 23%. 13% of the samples are neither subtype G nor CRF02_AG. Notably, 13 out of 15 recombinants contain subtype G gp120 sequences. Our data agree with previous reports showing a higher percentage of subtype G in southern Nigeria (Enugu) as compared to the north (Kaduna). Conclusion: Subtype G and subtype G-positive gp120 recombinants represent a significant portion of HIV-1 infections in Nigeria, suggesting that the total number of subtype G infections in Western Africa may be more of a burden than previously thought and should inform global vaccine designs.

Background: The results from the RV144 Thai vaccine trial have shown a modest effect in preventing HIV infection. However, the underlying mechanisms of protection remain to be identified. Developing vaccine homologous reagents for in vitro immunogenicity assays is a priority for the field. Here we present the development and characterization of vectors expressing functional CM244 envelopes (component of AIDSVAX B/E). Methods: CM244 env was isolated from different sources: infected A3R5 cells (AD), PBMC virus passaged once from 1989 (OR), later passaged PBMC (ec1, NW) and a synthetic gene (v.i.). Sequenced plasmids encoding these envs were tested for infectivity and site-directed mutagenesis was used to correct non-functional and poorly infectious envelopes and/or to change amino acids to match the immunogen sequence. Neutralization assays were performed in TZMbl cells using pseudoviruses (PV) and in PBMC using infectious molecular clones (IMC). Results: Among CM244 clones, OR was the best PV env candidate (highly infectious and nearly homologous to the vaccine insert) and was inserted into clade CRF01_AE and clade C Renilla Luciferase backbones to generate chimeric IMCs. The synthetic env matching the vaccine insert (v.i.) did not mediate infection and showed specific mutations, including some affecting the CD4 binding site. Sequence comparisons revealed that clones with different cellular origin had unique mutations; in particular, a three amino acid variation occurred in the V1/V2 region, affecting the recently identified peptide 49 that may be associated with cellular immune responses. Preliminary neutralization results showed no major differences between PVs despite these mutations. However, when PV were compared with IMCs, striking differences were observed, including two-log increases in sensitivity of IMC to HIV+ serum and monoclonal antibody b12. Conclusion: These data highlight the importance of development of IMC for use in primary cell assays, as evidenced by differences observed in neutralization when the same envelope is tested in different platforms.

AIDS Vaccine 2011

225

POSTERS

Posters
P20.09 LB

Topic 20: Viral Diversity

The Prevalence of Low and High Viral Load in a Cohort of African Volunteers During the First Year of Infection with HIV-1 Subtype A1, C and D
M.A. Price8, E. Ruzagira1, S. Lakhi2, E. Karita3, W. Kilembe2, E.J. Sanders4, A. Kamali1, O. Anzala5, M. Latka6, S. Allen7, P.N. Amornkul8, E. Cormier8, P. Fast8, J. Gilmour8
1 Medical Research Council/Uganda Virus Research Institute, Masaka, Uganda; 2Zambia-Emory HIV Research Project, Lusaka, Zambia; 3Projet San Francisco, Kigali, Rwanda; 4 Centre for Geographic Medicine Research Coast/KEMRI, Kilifi, Kenya; 5Kenya AIDS Vaccine Initiative/University of Nairobi, Nairobi, Kenya; 6Aurum Institute for Health Research, Rustenberg, South Africa; 7Rwanda Zambia HIV Research Group, Atlanta, GA, USA; 8International AIDS Vaccine Initiative, San Francisco, CA, USA

Background: In trials evaluating HIV vaccine efficacy, the potential influence of HIV-1 subtype on viral load (VL) during early HIV breakthrough infection may be important in vaccine evaluation. Methods: HIV-infected volunteers in Kenya, Rwanda, South Africa, Uganda, and Zambia were enrolled into a seroconverter cohort within one year of estimated date of infection (EDI). Plasma VL post-EDI was measured monthly for 3 months, quarterly until 24 months, and 6-monthly thereafter. Acute and early HIV infection was defined as 90 days and 91-365 days post-EDI, respectively. VL data were analyzed as log10-transformed data. For each period, high and low VL was defined as a mean VL5.0x106 and VL3.3x106 copies/mL, respectively. Only visits before ART initiation were included. Results: Of 463 volunteers, 177(38.2%) were infected with HIV1 subtype A1, 216(46.6%) with subtype C, and 70(15.1%) with subtype D. 407 volunteers had acute infection (AI): 158(38.9%) had subtype A1, 183(44.9%) had C and 66(16.2%) had D; 157(38.6%) had high VL and 42(10.3%) had low VL; neither varied by subtype (p=0.6 and 0.8, respectively). Median VL during AI(5.1x106copies/mL) was subtype-independent(p=0.3). During early infection (EI), 115/463(24.8%) volunteers had high VL, which did not vary by subtype (p=0.2). Low VL during EI was significantly associated with subtype A1(26/177, 14.7%) vs. C(13/216, 6.0%, p=0.01) but not associated with subtype A1 vs. D(p=0.1). Controlling for sex and infecting subtype, women were significantly more likely(OR 2.9, p=0.001) to have low VL during EI, while individuals with subtype C infection(OR 0.31, p=0.001) were significantly less likely. Median VL during EI, 4.5x106copies/ mL, did not vary by subtype(p=0.07). Conclusion: During early HIV infection in this African cohort, we observed a significant association between subtype A1 and low VL. The potential confounding effect of infecting HIV-1 subtype should be considered when evaluating HIV vaccine efficacy using viral load as surrogate endpoint in breakthrough infections.

POSTERS 226 AIDS Vaccine 2011

Topic 21: Virology and Immunology of HIV Transmission P21.01


Electrophoretic Fingerprint of HIV-1 Env Reveals Conformational Changes that Model Male to Female Transmission Events
D.J. Stieh1, D. Fairhurst2, S. Jeffs3, D. Montefiori4, C. Takawira1, R. Shattock1
1

Posters

P21.02
Epidemiology of Adenovirus Type 5 Neutralizing Antibodies in Healthy People and AIDS Patients in Southern China
S. Caijun1, Z. Yinfeng1, F. Liqiang1, P. Weiqi1, C. Ling1
1 Guangzhou Institutes of Biomedicine and Health, Guangzhou, China

St. Georges, University of London, London, United Kingdom (Great Britain); 2International Partnership for Microbicides, Bethesda, MD, USA; 3Division of Infectious Diseases, Imperial College, London, United Kingdom (Great Britain); 4 Department of Immunology, Duke University Medical Center, Durham, NC, USA
Background: Understanding of the biochemical nature of events that take place during HIV-1 transmission enables rational design of immunogens and understanding of the environment where vaccine responses must be efficacious. Electrophoretic mobility and binding kinetic measurements allow characterization of the surface charge and determination of affinity changes between Env and receptors. It is hypothesized that different surface chemistries can be maintained through chemical cross-linking which defines conditions for inducing more potent immune responses. Methods: Electrophoretic mobility and affinity for the CD4 molecule was measured for CN54 and BX08 gp140 trimers across a range of pH. The gp140 conformations present at varying pH values were fixed with gluteraldehyde and their immunogenicity and ability to induce neutralizing antibodies was assessed in rabbits. Intramuscular prime and boosts were administered at 4 week intervals with or without gp140 stabilizing molecule DS003 and adjuvanted with LASTS. Serum was collected and analyzed for native and immunogen specific IgG and neutralization in TZM-bl assays. Results: gp140 trimers demonstrated distinct profiles, whereby their mobility increases from pH 6.0 to 7.5. This coincided with decreased CD4 binding to gp140. Maximal binding occurred at pH 4.5, and the strongest affinity at pH 5.5. Immunization with protein fixed at pH 4.0 enhanced CN54 specific IgG induction, and neutralization titre increased against strain MW965.26. Other fixation conditions or the presence of DS003 did not change the neutralization profile. Conclusion: Env trimers from two different clades demonstrate increase in mobility as pH approaches neutral that is paralleled by decreased affinity for HIV-1 receptor CD4. Given that vaginal pH can range from 4-7, these data suggest that there are different conformations of Env present at mucosal pH ranges that influence transmission efficiency. Intramuscular immunization with the protein conformation present in these conditions enhanced neutralization against one sensitive strain and did not change response against tier 2 strains.

Background: Recombinant adenoviruses, especially serotype 5 (Ad5), have been extensively explored as vectors for vaccine or gene therapy. However, one major obstacle to their clinical applications is the high prevalence of preexisting immunity against adenoviruses resulting from natural infection. It has been reported that there are geographic variations in the prevalence of natural adenovirus infection. Methods: We investigated the seroprevalence of Ad5 in Guangzhou, southern China by measuring the preexisting Ad5 immunity by SEAP-based neutralizing assay. Results: The seroprevalence was 77.34% in general healthy population. The older population (age 41-72) had the highest seropositivity (84.8%) and the highest percentage (54.4%) having high antibody titers (>1000), while the younger population (age 12 or less) had the lowest percentage (30.6%) having high antibody titers (>1000). The dynamics of Ad5 neutralizing antibodies were stable and persistent over the course of eight months. In light of concerns that high Ad5 antibodies may be associated with increased susceptibility to HIV infection, we also investigated the seroprevalence of Ad5 in the HIV-infected AIDS patients in southern Chinaand our data demonstrate that there was not a significant correlation between Ad5 neutralizing antibodies titers with HIV infection. Conclusion: Our study provided useful insights for the future development of Ad5-based HIV vaccine and gene therapy.

AIDS Vaccine 2011

227

POSTERS

Posters
P21.03

Topic 21: Virology and Immunology of HIV Transmission P21.04


Drug Resistance Pattern of HIV-1 Among TherapyNaives in North-Central Nigeria Suggests Regional Differences in Transmitted Drug Resistance Rates
H.O. Ajoge1, M.L. Gordon2, S. Ibrahim3, O.S. Shittu4, T. Ndungu2, S.O. Olonitola1
1

Human Immunodeficiency Virus Type 1 Variants Issued from Mother-Child Pairs Display a Wide Spectrum of Biological Properties
S. Thenin1, M. Braibant1, T. Samleerat2, N. Ngo-Giang-Huong3, G. Jourdain3, M. Lallemant3, F. Barin1 Universit F. Rabelais. INSERM U966, Tours, France; 2Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand; 3Harvard University-Institut de Recherche pour le Dveloppement,UMI174, Chiang Mai, Thailand
1

Background: Mother-to-child transmission is the leading source of HIV infection in children. Several studies have demonstrated that early virus population present in infected infants usually is homogeneous when compared to viral population of mothers at delivery. It has been shown also that variants in infants are frequently resistant to neutralization by autologous maternal plasma. In this study, we analyzed biological functions of envelope genes from two mother-infant pairs. Methods: We explored antigenic and functional properties of pseudotyped viruses expressing gp120 encoded by several env clones issued from two mother-infant pairs (0978 and 1021) infected by CRF01_AE viruses. We compared their sensitivity to neutralization by autologous and heterologous sera and by neutralizing monoclonal antibody b12. We also analyzed celltropism, sensitivity to sCD4 and to the CCR5 antagonist TAK-779, and infectivity in TZM-bl cells. Results: We obtained infectious viruses from three mothers env clones and three infants env clones from pair 0978, and from one mothers env clone and two infants env clones from pair 1021. All nine clones exhibited CCR5 tropism, had a high sensitivity to heterologous neutralization, were resistant to b12 and presented equivalent sensitivity to TAK-779. However, sensitivity to sCD4, and infectivity were highly heterogeneous within both pairs and cases. All clones were resistant to autologous neutralization, except a single clone issued from infant 0978 (0978-E2) which presented a high sensitivity to maternal plasma. By site-directed mutagenesis we showed that a single mutation Valine/Alanine at position 68 in the C1 region governed this sensitivity to autologous neutralization and was also involved in sensitivity to sCD4 and infectivity. Conclusion: This study showed the already heterogeneous functional properties of env genes from infected infants despite a homogeneous genetic virus population early after infection, and demonstrated the possible impact of a specific C1 residue in both sensitivity to autologous neutralization, sensitivity to sCD4 and viral infectivity.

Department of Microbiology, Ahmadu Bello University, Zaria, Nigeria; 2HIV Pathogenesis Programme, DDMRI, University of KwaZulu-Natal, Durban, South Africa; 3Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; 4 Department of Gyneacology and Obstetrics, ABUTH, Ahmadu Bello University, Zaria, Nigeria
Background: In Nigeria, the country with the second largest number of HIV-1 infected people globally, antiretroviral therapy rollout is now widespread with an increasing number of individuals and communities benefitting. However, the drug resistance profile of patients initiating or failing on antiretroviral therapy as well as transmitted drug resistance (TDR) rates is not well characterized; especially in North-Central Nigeria (NCN). NCN is one of Nigerias six geopolitical zones and NCNs HIV prevalence has remained the highest over time. Methods: Molecular variability of the protease and reverse transcriptase region of isolates from therapy-naive pregnant women sampled in 2007 from NCN was studied by amplifying, cloning and sequencing DNA after reverse transcription of plasma RNA. Nucleotide sequences were translated to amino acid sequences and submitted to the Stanford HIV Drug Resistance Database to identify drug resistance mutations/polymorphisms. Results: Of 28 samples amplified, 14, one, one, one and 11 were found to be concordantly CRF02_AG, subtypes C, F2, and G respectively, in both genes. Among various Protease mutations/ polymorphisms, high prevalence of K20I/R (27/28), M36I (28/28) and V82I (all subtype G samples) was observed. Contrary to observation from other geopolitical zones, lesser frequency of reverse transcriptase mutations/polymorphisms was observed. Conclusion: As in previous studies on therapy-naives from other geopolitical zones, the predominance of CRF02_AG and subtype G as well as presence of both primary and secondary drug resistance mutations was observed. However the lesser frequency of reverse transcriptase mutations/polymorphisms, suggests differences in level of TDR in the different geopolitical zones (previous being from South-West with older/wider antiretroviral therapy rollout). Since the prevalence of NRTI and NNRTI TDR is largely unknown in Nigeria (unlike in the developed countries where the TDR rates are known), we recommend surveillance studies on TDR in Nigeria in order to monitor the now widespread antiretroviral therapy rollout.

POSTERS 228 AIDS Vaccine 2011

Topic 21: Virology and Immunology of HIV Transmission P21.05


Lack of Association Between HSV-2 and HIV-1 Infection in a Cohort of HIV-1 Discordant and Concordant Couples in Dakar, Senegal
M. Camara1, T.N. Dieye1, M. Seydi2, P.S. Sow2, S. Mboup1, L. Kestens3 Laboratory of Immunology, Aristide Le Dantec University Hospital, Dakar, Senegal; 2Department of Infectious Diseases, CHU Fann University Hospital, Dakar, Senegal; 3Laboratory of Immunology, Department of Microbiology, Institute of Tro, Antwerp, Belgium
1

Posters

P21.06
Role of Antibody and Complement in Neutralization of Trans Infection by Erythrocytebound HIV-1
Z. Beck1, B.K. Brown1, G.R. Matyas2, V.R. Polonis2, M. Rao2, C.R. Alving2
1

Henry M Jackson Foundation / U.S. Military HIV Research Program, Rockville, MD, USA; 2U.S. Military HIV Research Program, Walter Reed Army Institute of Rese, Rockville, MD, USA

Background: A synergy between HIV-1 and HSV-2 infections has been reported in numerous studies. However, recent clinical trials testing the efficacy of HSV-2 suppressive acyclovir therapy failed to show an effect on HIV-1 acquisition and transmission. Methods: In this study, we reassessed the putative association between HSV-2 and HIV-1 infection in a cohort of HIV-negative, HIV-1 discordant and HIV-1 positive concordant couples in Dakar, Senegal. Results: In agreement with previous studies, we observed a strong overall association between HSV-2 and HIV-1 serostatus (OR, 4.61; P < 0.001). However, this correlation appeared to be driven by a low HSV-2 prevalence in HIV-negative couples relative to HIV-discordant and concordant couples (23% vs 59% and 66% of couples with at least one HSV-2 positive partner, respectively; P < 0.001). HIV-discordant and concordant couples showed comparable frequencies in HSV-2 seroprevalence (59% vs 66%; P = 0.483), and differences in HSV-2 status among index (59% vs 62%, P = 1.000) and recipient partners (63% vs 41%, P = 0.131) between the two groups were absent or insufficiently large to suggest a causal relationship between HSV-2 and HIV-1. Conclusion: The interaction between HIV and HSV-2 appears complex, and precise longitudinal studies are required to dissect their exact temporal relationship.

Background: In neutralization assays, free virus is generally used as the infectious virus source. It has recently been observed that HIV-1 can bind to human erythrocytes, and that erythrocytebound HIV-1 remains infectious and promotes trans infection of CD4(+) T cells in vitro. In neutralization assays, plasma (or serum) as the antibody source, is pre-heated to inactivate complement (C). However, innate effector mechanisms can be important for prevention of HIV-1 infection, and also be utilized by HIV-1 to promote virus entry. Methods: We have recently studied the ability of two broadly neutralizing human monoclonal IgG antibodies, 4E10 and b12, to prevent trans infection of CD4(+) PBMC by erythrocyte-bound HIV-1 both in the presence and absence of complement. The virus source consisted of either cell-free HIV-1 for direct infection, or HIV bound to erythrocytes for trans infection of permissive cells. Results: In the absence of C mAbs neutralized trans infection of erythrocyte-bound virus less effectively than cell-free virus. At a low concentration, 4E10 enhanced trans infection. In the case of 4E10, antibody-dependent C activation inhibited trans infection by erythrocyte-bound HIV-1, but caused enhanced infection with cell-free HIV-1 in the presence of erythrocytes. No effects of C were observed with b12. Conclusion: Complement-independent enhancement is proposed as due to Fc-receptor mediated uptake of the virus by the PBMC, and complement-dependent enhancement in the presence of erythrocytes is proposed as due to binding of C3b-4E10-cellfree-HIV or C3d-4E10-cell-free-HIV to C receptor type 1 on erythrocytes, or C receptor type 2 on B cells in the PBMC. We conclude that erythrocytes may serve as an immunologically protected site for infectious HIV-1 virions that can cause HIV-1 trans infection of susceptible cells. We also conclude that antibody-dependent activation of C may cause either neutralization or enhancement of trans infection of PBMC by erythrocyte-bound HIV-1 depending on C activation.

AIDS Vaccine 2011

229

POSTERS

Posters
P21.07

Topic 21: Virology and Immunology of HIV Transmission P21.08


The Removal of Transmission-linked Glycosylation Sites in HIV gp120 Promotes High a4b7 Reactivity
C. Cicala1, D. Van Ryk1, F. Nawaz1, M. Pascuccio1, K. Jelicic1, D. Wei1, N. Patel1, K. Block1, J. Arthos1, A.S. Fauci1
1

Influence of the Menstrual Cycle on Levels of Innate Immune Proteins in the Genital Tract of Female Pigtail Macaques
A. Vishwanathan1, P.C. Guenthner1, R. Hendry1, J.M. McNicholl1, E.N. Kersh1 Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
1

National Institute of Allergy and Infectious Diseases (USA), Bethesda, MD, USA

Background: Mucosal innate factors from the female reproductive tract (FRT) might be critical in modulating HIV-l transmission. Most of the innate factors are known to be influenced by sex hormones, shown in humans but not extensively studied in pigtail macaques. Considering that pigtails, like humans, exhibit continuous lunar menstrual cycles, and are important models for HIV-1 transmission, we examined levels of various innate proteins in the context of their menstrual cycles. We hypothesized that certain immune proteins would be up-regulated in the follicular phase of the cycle compared to the luteal phase, based on our previous demonstration of increased susceptibility to vaginal SHIV infection in the luteal phase. Methods: Blood and cervico-vaginal lavages were collected weekly over twelve weeks from ten untreated, uninfected adult female pigtails. In addition to measuring plasma progesterone, innate immune factors [trappin-2, secretory leukocyte protease inhibitor (SLPI), thymic stromal lymphopoietin, surfactant protein-D, defensins], cytokines (IL-1, IL-6, IL-8, TNF-, G-CSF), and chemokines (RANTES, eotaxin, MCP-1) were quantified in vaginal fluids by ELISA or Luminex technology. For this analysis, the beginning of a menstrual cycle in each animal was defined as the time point following steepest decline of progesterone; statistical significance was calculated using Mann-Whitney nonparametric test. Results: Four proteins showed significantly higher levels in the follicular phase, including TNF- (p=0.0079), G-CSF (p=0.0205), MCP-1 (p=0.0041), and Eotaxin (p=0.0349). The levels of remaining proteins, including SLPI (p=0.1918), did not exhibit significant differences based on the phase of the menstrual cycle. Conclusion: Four mucosal innate factors were up-regulated in the follicular phase which is also the phase of lowered susceptibility to infection. This indicates that pigtail macaques have similarly regulated mucosal immune factors in the FRT as observed in women during the menstrual cycle. This study highlights the suitability of female pigtails as a model for female sexual HIV-1 acquisition.

Background: Mucosal transmission of HIV is typically inefficient. The virus must breach physical barriers before it infects mucosal CD4+ T cells. Low-level viral replication occurs initially in mucosal CD4+ T cells, but within days high-level replication occurs in Peyers patches, mesenteric lymph nodes and the gut lamina propria. Early-replicating viruses in subtypes A and C mucosal transmission tend to encode gp120s with reduced numbers of N-linked glycosylation sites at specific positions throughout the V1- V4 domains, relative to typical chronically replicating isolates. The selective advantage gained by the absence of these N-linked glycosylation sites is unknown. Methods: Using primary a4b7+/CD4+ T cells and a flow-cytometry based steady-state binding assay we show that the removal of N-linked glycosylation sites associated with early stages of infection in chronic subtype A and C gp120s results in large increases in the specific reactivity of gp120 for integrin-a4b7. Results: Next we followed infection longitudinally in individual patients and found that high-affinity for integrin a4b7reactivity, although not found in only some chronic gp120s, was observed in the early-transmitting gp120s that we analyzed. Early-transmitting gp120s were further distinguished by their dependence on avidity-effects to interact with CD4, suggesting that gp120s with high affinity for a4b7+/CD4+ T cells bear unusual structural features not present in typical chronic gp120s. Conclusion: These results suggest that at an early stage following transmission there exists a requirement for the productive infection of a4b7+/CD4+ T cells. Understanding the structural features that characterize early-transmitting gp120s and the role of integrin a4b7 receptor in HIV transmission fitness, may thereby aid in the design of novel vaccine immunogens.

POSTERS 230 AIDS Vaccine 2011

Author Index
A
Abaasa, A . . . . . . . . . . . P15.07 Abagyan, R . . . . . . . . . . P05.05 Abdool Karim, Q . . . . . . OA01.04 Abdool Karim, S . . . . . . . OA01.04, OA04.07 LB, OA06.04, P04.03, P04.24, P04.29, P08.02, P17.20 Abdulhaqq, SA . . . . . . . . . P14.11 Abisa, J . . . . . . . . . . . P15.13 LB Aboud, S . . . . . . . . . . . . P14.06 Aboulker, J . . . . . . . . . . P14.01 Abrahams, F . . . . . . . . . . P04.28 Abrahams, M . . . . . . . . . P17.20 Ackland, J . . . . . . . . . . OA09.01 Adam, L . . . . . . . . . . . . P17.23 Adams, DJ . . . . . . . . . . OA02.01 Adams, E . . . . OA01.02, OA01.06 LB Addo, M . . . . . . OA05.06, P01.04, P09.13 Aderem, A . . . . . . OA02.01, S05.05 Adeyemi, A . . . . . . P15.10, P16.02, P16.05 Adler, M . . . . . . . . . . . OA10.03 Afolabi, M . . . . . .OA09.06, P12.02 Agboola, S . . . . . . P16.01, P16.02, P16.05 Ahmad, AA . . . . . . . . . . P20.05 Ahmad, F . . . . . . . . . . . P09.05 Ahmed, FK . . . . . . . . . . . P19.29 Aidarus, N . . . . . . . . . P18.24 LB Ajoge, HO . . . . . . .P20.05, P21.04 Akahata, W . . . . . . . . . . PL03.01 Akahoshi, T . . . . . . . . . . P17.08 Ake, J . . . . . . . . . . . . OA01.02 Akiyama, H . . . . . . . . . . P04.20 Akulova, E . . . . . . . . . . . P14.09 Alam, SM . . . . . . . . . . P04.39 LB Alcam, J . . . . . . . . P11.04, P11.05 Alcena, DC . . . . . . . . . . . P04.19 Aldebert, D . . . . . . . . . . . P19.11 Alexandre, KB . . . . . . . . . P15.04 Alfai, E . . . . . . . . . . . . . P06.09 Alicea, C . . . . . . . . . . . OA10.04 Allen, S . . . . P04.41 LB, P06.14 LB, P06.16 LB, P08.01, P20.09 LB, S06.01 Allen, T . . . . . . OA02.02, OA03.02, OA05.06, P08.03, P09.02, P09.07, P09.08, P17.15 Almeida, DV . . . . . . . . . . P04.08 Almeida, RR . . . . . .P18.13, P18.18 Almond, N . . . . .P17.28 LB, P18.15, P18.17 Alpert, MD . . . . . .OA09.02, P19.27 Alter, G . . . . P04.11, P09.13, S03.04 Altfeld, M . . . . OA01.04, OA02.02, OA03.02, OA05.02, OA05.06, P09.01, P09.02, P09.07, P09.08, P09.13, P17.15, S03.04 Alvarez-Fernndez, CC . . . . P11.04, P11.05 Alving, CR . . . . . OA02.05, P02.06, P02.07, P21.06 Amacker, M . . . . . . . . . OA10.03 Amara, R . . . . . OA01.05, OA10.04, P02.08 Ambler, G . . . . . OA09.06 , P12.02 Ameresena, T . . . . . . . . OA08.07 Ami, Y . . . . . . . . . . . . . P18.04 Amornkul, PN . . . . . . . . . P04.09, P20.09 LB Ananworanich, J . . . . . . . P01.03, P10.07 LB Andersen, H . . . . . . . . . OA08.04 Andersen, P . . . . . . . . . . P13.01 Andersen-Nissen, E . . . . . OA02.01 Anderson, M . . . . . . . . . OA08.03 Andersson, S . . . . . P06.09, P06.10 Andrabi, R . . . . . . . . . . . P04.17 Andrasik, M . . . . . . . . . . P15.11 Andreas, B . . . . . . . . . . . P14.06 Andrews, CD . . . . . . . . . . P02.04 Angin, M . . . . . . . . . . . P01.04 Anglister, J . . . . . . . . . . . P04.15 Anjuere, F . . . . . . . . . . OA10.03 ANRS COHVAC Study Group and the ANRS HIV Vaccine Network/VRI . . . . . . . . . P14.01 Ansari, AA . . . . . . . . OA02.06 LB Anzala, AO . . . . P20.09 LB, S04.05 Arberas, H . . . . . . . P09.10, P11.04 Ardito, M . . . . . . . P18.19, P18.20 Arias, M . . . . . . . . . . . . P10.04 Aris, EA . . . . . . . . . . . . P14.06 Arnold, V . . . . . . . . . . OA02.03 Arrode-Brusses, G . . . . . . . P19.11 Arroyo, M . . . . . . . . . . . P20.04 Arshava, B . . . . . . . . . . . P04.15 Arthos, J . . . . . . . . P21.08, S06.02 Arworn, D . . . . . . . . OA07.08 LB Asher, L . . . . . . . OA02.05, P02.06 Ashraf, A . . . . . . . . . . P17.29 LB Asiki, G . . . . . . . . . . . . P15.05 Asmuth, D . . . . . . . . . P18.21 LB Assawadarachai, V . . . . . P10.07 LB, P20.04 Astanina, M . . . . . . . . . . P14.09 Asti, V . . . . . . . . . . . . . P11.03 Athanasopoulos, T . . . . P17.33 LB, P18.17, P18.23 LB Atman, P . . . . . . . . . . . P19.01 Autran, B . . . . . . . . . . . P19.08 Avila, C . . . . . . . . . . . . P16.08 Avila, MM . . . . . . .P15.01, P15.02 Avila-Rios, S . . . . . . . . P07.04 LB Axten, K . . . . . . OA02.02, P09.02, P09.07 Axthelm, M . . . . . . . . . . P10.05 Ayemoba, OR . . . . . . . P20.07 LB Azeez, A . . . . . . . . . . . . P15.10 Azzoni, L . . . . . . . . . . . . P14.11 Ball, TB . . . . . . P07.03 LB, P17.03 Balla-Jhagjhoorsingh, SS . . P04.35 LB Baltimore, D . . . . . . . . . OA04.06 Ballmaier, M . . . . . . . . . . P09.05 Banchereau, J . . . OA03.01, P01.06, P10.10 LB, S05.02 Bansal, A . . . . . . . . . . . P17.06 Bar, K . . . . . . . . . . . . . S07.03 Barbagalo, R . . . . . . . . . . P18.17 Barin, B . . . . . . . . . . . OA09.01 Barin, F . . . . . . . . . . . . P21.03 Barnett, S . . . OA03.05 LB, OA10.06, P05.07, P19.33 LB Baron, M . . . . . . . . . . . . P14.08 Barouch, D . . . . . OA11.04, P05.03, P10.08 LB, P14.05, P17.13, P17.14, P19.30 LB, S06.05 Barr-Sinoussi, F . . . . . . OA02.03 Barreda, V . . . . . . . . . . . P15.02 Bart, P . . . .OA01.01, P17.11, P20.01 Bates, A . . . . . . . . . . . . S07.01 Bayingana, R . . . . . . . P06.13 LB, P06.15 LB, P06.16 LB Bean, AT . . . . . . . . . . . OA08.02 Beck, Z . . . . . . . . . . . . . P21.06 Becker, P . . . . . .P17.33 LB, P18.17, P18.23 LB Beckett, T . . . . . . . . . . . P05.07 Beenhakker, N . . . . . . . . . P18.05 Behrendt, R . . . . . . . . . . P05.02 Belisle, SE . . . . . . . . . . OA08.04 Bellier, B . . . . . . . .P19.05, P19.08 Bello, G . . . . . . . . P04.08, P20.03 Bellutti Enders, F . . . . . . . . P17.11 Beloqui, J . . . . . . . . . . . P16.10 Benenson, M . . . . . P01.07, P06.05, P14.03 Benfield, T . . . . . . . . . . . P17.19 Benlahrech, A . . .P17.33 LB, P18.17, P18.23 LB Bere, A . . . . . . . . . . . . . P10.02 Berenberg, J . . . . . . . . . . P14.03 Berkhout, B . . . . . . . . P17.28 LB Berman, P . . . . . . .P04.33, P04.34 Bernard, NF . . . . . . . . . OA02.04 Bernardo, ML . . . . . . . . . OA11.03 Berry, M . . . . . . . . P01.06, S05.02 Berry, N . . . . . . . . . . . P17.28 LB Bess, Jr., J . . . . . . . . . . OA10.04 Bet, A . . . . . . . . . . . . . P17.06 Bett, A . . . . . . . P14.14 LB, P19.20 Betts, M . . . . . . . . . . . . P19.24 Bewley, C . . . . . . . . . . OA03.03 Beyrer, C . . . . . . . . . . . . P06.04 Bhatnagar, N . . . . . . . . . P09.05 Bhatt, N . . . . . . . . . . . . P06.10 Bhiman, J . . . . OA04.07 LB, P04.03, P04.29, P08.02 Bhoumik, P . . . . . . . . . . P17.15 Bian, B . . . . . . . . . . . . . P19.01 Biberfeld, G . . . . OA09.03, P06.10, P14.04, P14.06 Bibert, S . . . . . . . . . . . . P19.20 Bickel, M . . . . . . . . . . . OA03.04 Biedma, ME . . . . . . . . . . P04.26 Bielawny, T . . . . . . P17.03, P18.12 Billings, E . . . . OA07.08 LB, OA09.04 Billingsley, JM . . . . . . . . . P03.01 Binello, N . . . . . . . . . . . P07.02 Binley, J . . . P04.07, P05.01, P19.03 Birks, J . . . . . . . . . . . . . P17.16 Bishop, K . . . . . . . . . . OA05.02 Black, A . . . . . . .OA09.06, P12.02 Blake, TB . . . . . . . . . . . . P09.09 Blankenship, D . . . . P01.06, S05.02 Blay-Puryear, W . . . . . . . . P05.07 Bleiholder, A . . . . . . . . . . P05.02 Block, K . . . . . . . . . . . . P21.08 Blume-Peytavi, U . . . . . . . P19.08 Boente-Carrera, M . . . . . . P02.04 Boeser-Nunnink, BD . . . . . P04.06 Bogers, W . . . . . . .P04.04, P18.05 Bolen Valenzuela, A . . . . . . P01.07 Boliar, S . . . . . . . . . . P04.41 LB Bomsel, M . . OA10.03, P11.03, S02.01 Bongertz, V . . . . . . . . . . P04.08 Bonsignori, M . . OA03.03, P04.36 LB, P04.39 LB, P04.40 LB Borrow, P . . . . . . . . . . . P08.01 Borthwick, N . . . . OA09.06 , P12.02 Bosch, RJ . . . . . . . . . . . P09.01 Bose, M . . . . . . . . . . . . . S07.01 Bosquet, N . . . . . . .P17.23, P19.16 Boutwell, CL . . . . . . . . . OA03.02 Bouzek, H . . . . . . . . . . . S07.01 Bowles, EJ . . . . . . . . . . . P04.25 Bowornwatanuwong, J . . OA01.06 LB, P14.13 LB Boyer, J . . . . . . OA01.02, OA08.04 Boyington, J . . . . OA03.03, PL03.01 Boyle, CM . . . . . . . . . . . P18.19 Bradfield, A . . . . . . . . P20.08 LB Bradley, RR . . . . . . . . . . P17.13 Bradfield, A . . . . . . . . . . S07.01 Braibant, M . . . . . . . . . . P21.03 Brander, C . . . . . . P06.06, P17.24, P19.08, P19.22 Brandt, L . . . . . . . .P13.01, P17.19 Bravo, I . . . . . . . . . . . . P06.06 Brenchley, J . . . . .OA08.01, P17.22 Bridgeman, A . . . . . . . . OA09.06 Brill, B . . . . . . . . . . . . OA03.04 Brinckmann, S . . . . . P02.09, P02.10 Briones, M . . . . . . . . . . . P09.04 Brockman, MA . . . . . . . . P08.04 Broder, G . . . . . . . . . . . . P15.11 Broderick, K . . . . . . . . . . P19.01 Brooks, A . . . . . . . . . . OA08.07 Brown, BK . . OA07.06, P04.13, P21.06 Brown, J . . . . . . . . . . . . P01.02 Brumme, CJ . . . . . . . . . . P08.04 Brumme, ZL . . . . . . . . . . P08.04 Buapunth, P . . . . . . . . . . P01.07 Buchbinder, S . . . . . . . P18.27 LB, P19.30 LB, S07.01 Buck, CB . . . . . . . . . . . OA11.03 Budde, ML . . . . . . . . . P17.31 LB Buffa, V . . . . . . . . P02.01, P10.03 Buggert, M . . . . . . . . . . P17.09 Bulmer-Thomas, B . . . . . . P04.14 Buma, D . . . . . . . . . . . . P14.06 Bunce, C . . . . . . .OA09.01, P15.08 Bunce, RN, MS, CA . . . . . . P06.08 Bunjun, R . . . . . . . . . . . . P11.07 Bunnik, EM . . . . . . . . . . P04.06 Buonaguro, FM . . . . P11.03, P19.15 Buonaguro, L . . . . . P11.03, P19.15 Burger, JA . . . . . . . . . . . P04.06 Burgers, WA . . . P04.28, P10.02, P11.07 Burton, DR . . . . OA04.01, OA04.05, OA10.05, P04.27, P04.42 LB, P04.40 LB, P05.06, P19.29, S02.05

Bachy, V . . . . . . . . . . . . P18.17 Baden, LR . . . . . . . . . P10.08 LB Bagarazzi, M . . . . . . . . OA01.02 Bahemuka, U . . . . . . . . . P15.07 Bailer, R . . . . . . . . . . . . P04.27 Bailey, M . . . . . . . . . . P17.32 LB Bailly-Bechet, M . . . . . . . P09.12 Bakari, M . . . . . . . . . . . P14.06 Baker, A . . . . . . . . . . . . S01.01 Baksaas, I . . . . . . . . . . P18.21 LB Bala, M . . . . . . . . . . . . P04.17 Balan, I . . . . . . . . . . . . P15.02 Balazs, AB . . . . . . . . . . OA04.06 Baldwin, N . . . . . . . P01.06, S05.02 Ball, B . . . . . . . . . . . . . P18.12 Ball, T . . . . . . . . . P17.04, P17.05

AIDS Vaccine 2011

231

AUTHOR INDEX

Author Index
Burwitz, BJ . . . . . . . . . P17.31 LB Busari, A . . . . . . . .P16.01, P16.05 Busari, OA . . . . . . .P16.01, P16.05 Busaro, OA . . . . . . . . . . P16.02 Bussaratid, V . . . . . . . . . P14.03 Bwanika, A . . . . . . . . . . P15.07 Bychenko, A . . P03.02 LB, P19.31 LB Cheeseman, H . . . . . . . P17.29 LB Chege, G . . . . . . . . P11.07, P18.11 Chen, B . . . . . . . . . . . . P05.03 Chen, H . . . . . . . . . . . . P18.02 Chen, J . . . . . . .OA04.06, OA06.06 Chen, L . . . . . . . . P04.14, P15.06, P19.12, P19.17 Chen, RT . . . . . . . . . . . . P16.04 Chen, W . . . . . . .OA04.03, P14.02 Chen, X . . . . P04.36 LB, P04.37 LB, P04.39 LB Chen, Y . . . . . . . . . . . . P04.31 Chen, Z . . . . P15.06, P19.12, P19.17 Chene, G . . . . . . . . P01.06, S05.02 Cheng, H . . . . . . . . . OA03.05 LB Chenine, A . . . . . . . . . P20.08 LB Chetty, S . . . . . . . . . . . . P17.01 Chibnik, LB . . . . . . . . . OA06.01 Chikwamba, R . . . . . . . . . P15.04 Chin, EN . . . . . . . . . . P17.31 LB Chiu, J . . . . . . . . . . . . . P14.03 Cho, M . . . . . . . . . . . . OA10.01 Chomba, E . . . . . . . . . P06.14 LB Chomchey, N . . . . . . . . P10.07 LB Chonco, F . . . . . . . . . . OA05.02 Chonwattana, W . . . P06.12, P20.04 Chopera, DR . . . . .OA06.04, P08.04 Chow, S . . . . . . . . . . . . P09.04 Chua, B . . . . . . . . . . . . . P11.01 Chung, A . . . . . . OA06.05, P04.05 Chunsutthiwat, S . . . . OA01.06 LB Chunsuttiwat, S . . . . . . . . P14.03 Churchill, MJ . . . . . . . . . . P11.02 Churikanont, N . . . . . . P14.13 LB Ciani, G . . . . . . . . . . . . S06.01 Cicala, C . . . . . . . . . . . . P21.08 Cisper, N . . . . . . . . . . . . P19.24 Cisto, J . . . . . . . . . . . P19.33 LB Clark, B . . . . . . . . P04.32, P19.29 Clark, S . . . . . . . . . . . . OA11.04 Clausen, LN . . . . . . . . . . P17.19 Clement, F . . . . . . . . . . OA10.03 Clotet, B . . . . . . . . . . . . P06.06 Coalter, V . . . . . . . . . . OA08.06 Coetzee, D . . . . . . . . . . OA11.05 Cohen, M . . . . . . . P01.06, P04.24, S05.02 Cohen, S . . . . . . . . . . . OA10.02 Cole, AM . . . . . . . . . . P15.12 LB Coll, P . . . . . . . . . . . . . P06.06 Colleton, B . . . . . . . . . . P19.06 Collier, A . . . . . . . . . . . . P01.01 Coly, AA . . . . . . . . . . P17.30 LB Combadiere, B . . . . . . . . . P19.08 Conant, MA . . . . . . . . . . P04.33 Cong, Z . . . . . . . . . . . OA08.05 Connole, M . . . . . OA11.02, P03.01 Connors, M . . . . OA07.03, OA08.06, P04.42 LB Contreras, V . . . . P10.10 LB, P14.08 Coombes, N . . . . . . . . . . P10.05 Cooper, DA . . . . . . . . . P17.32 LB Coovadia, AH . . . . . . . . . P09.11 Cope, A . . . . . . . . . . . . P19.19 Corey, L . . . . P18.27 LB, P18.28 LB, S07.01 Cormier, E . . . . . OA09.01, P04.09, P08.01, P20.09 LB Cornara, S . . . . . . . . . . . P07.02 Crtes, F . . . . . . . .P04.08, P20.03 Cortez, VC . . . . . . . . . . OA07.07 Corti, D . . . . . . . . . . . P04.35 LB Cosma, A . . . . . . . P14.08, P17.23 Costagliola, D . . . . . . . . . P19.08 Cox, J . . . . . . . . OA09.01, P06.03, P14.12 LB, P17.29 LB, P18.06 Cranage, MP . . . . . . . . P17.28 LB Creegan, M . . . . . . . . . . P01.05 Crooks, ET . . P04.07, P05.01, P19.03 Crump, JA . . . P04.39 LB, P04.40 LB Cuburu, N . . . . . . . . . . OA11.03 Cummings, J . . . . . . . . . OA02.03 Cunha-Neto, E . . . . P18.13, P18.18 Curlin, M . . . . . . . . . . . P20.04 Curran, RM . . . . . . . . . . P10.06 Currier, J . . . . . OA01.02, OA05.03, OA07.06, OA07.08 LB, OA09.04, P14.05, P18.06 Cutler, S . . . . . OA05.01, OA05.06, P17.12 Cuzin, L . . . . . . . . . . . . P14.01 Czarnecki, C . . . . . . . . . . P18.12 Denny, T . . . . . . . . . . . . P06.03 Denny, TN . . . . . . . P01.06, S05.02 Derdeyn, C . . . . OA08.01, P04.41 LB, P08.01 Dereuddre-Bosquet, N . . P10.10 LB, P14.08 DeRosa, S . . . . . OA02.01, OA06.03 Desaint, C . . . . . . . . . . . P14.01 DeSilva, V . . . . . . . . . P14.14 LB Desjardins, D . . . . . . . . . P19.05 Desrosiers, RC . . . . . . . . . P19.27 Detmers, J . . . . . . . . . . OA10.03 DeVico, A . . . . . . . . . . . S02.02 DeVol, EB . . . . . . . P01.06, S05.02 Dewhurst, S . . . . . . . . . . P04.19 Dey, AK . . . . . . . . . . . P19.33 LB Dhalla, S . . . . . . . .P06.01, P06.02 Dhitavat, J . . . . . . . . . . . P14.03 Di Narzo, AF . . . . . . . . . . P20.01 Diallo, T . . . . . . . . . . . . P17.03 Diaw, P . . . . . . . . . . . P17.30 LB Dickson, G . . . . .P17.33 LB, P18.17, P18.23 LB Didier, C . . . . . . . . . . . OA02.03 Dieffenbach, C . . . . . . . . OC01.01 Dietrich, J . . . . . . . . . . . P16.03 Dietrich, U . . . . . . . . . . OA03.04 Dieye, TN . . . . . . . . . . . P21.05 Dimitrov, DS . . . . . . . . . OA04.03, P04.01, P14.02 Ding, X . . . . . . . . . . . . . P15.06 Diomede, L . . . . . . . . . . . P11.03 DiPasquale, J . . . . . . . . P14.14 LB Distefano, D . . . . . . . . . . P17.15 Division of AIDS (DAIDS) HIV Vaccine Trials Network . P15.11 Dolezal, C . . . . . . . . . . . P15.02 Dolin, R . . . . . . . . . . . P10.08 LB Doms, RW . . . . . . . . . . . P19.28 Donnelly, RF . . . . . . . . . . P10.06 Donofrio, G . . . . . . . . . . P01.05 Doores, KJ . . . . . .OA04.05, S02.05 Dor, V . . . . . . . . . . . . . P14.01 Dorfman, JR . . . . . . . . . . P04.28 Doria-Rose, N . . OA07.03, OA08.06, P04.27, P04.42 LB, P05.07 Doris, T . . . . . . . . . . . OA01.02 Dorrell, L . . . . . . . . . . . . P17.16 Dorta, G . . . . . . . . . . . OA01.01 Dossett, E . . . . . . . . . . . . P06.11 Doster, M . . . . . . OA01.03, P19.25 Douglass, N . . . . . . . . . . P18.11 Doyle, E . . . . . . . .P09.01, P09.02 Drillet, A . . . . . . . . . . . OA10.03 Drinker, M . . . . . . . . . . . P09.15 Du, Y . . . . . . . . . .P19.12, P19.17 Duan, D . . . . . . . . . . . . P18.09 Duan, L . . . . . OA11.01, OA11.06 LB Duchet-Niedziolka, P . . . . . P14.01 Dudek, TE . . . . . . OA03.02, P17.15 Duerr, A . . . . . . . OA02.01, P10.05 Durier, C . . . . . . . . . . . . P14.01

AUTHOR INDEX

C. Guardo, A . . . . . P09.10, P11.04, P11.05, P14.10 Cahn, P . . . . . . . . P17.02, P17.07 Caijun, S . . . . . . . P17.26, P19.10, P21.02 Calamante, G . . . . . . . . . P18.07 Calamel, A . . . . . . . . . . . P19.16 Calandra, T . . . . . . P19.18, P19.20 Calazans, G . . . . . . . . . . P16.10 Camara, M .P09.06, P17.30 LB, P21.05 Cameron, M . . . . . . . . . . S05.04 Capiga, SH . . . . . . . . . P04.39 LB Capina, R . . . . . . . P17.03, P18.10, P18.12 CAPRISA Study Team . . . . PL02.02 CAPRISA 002 Study Team . OA06.04, P17.20 CAPRISA 004 Trial Group . . OA01.04 Carballo-Dieguez, A . . P15.01, P15.02 Cardoso, SW . . . . . . . . . . P20.03 Cardozo, T . . . . OA09.05, OA10.02, P05.05 Carfi, A . . . . . . . . . . . P19.33 LB Carias, A . . . . . . . . . . . OA08.03 Carlson, J . . . . . . . P08.04, P17.06 Carnathan, D . . . . . . . . . P19.24 Carnathan, P . . . . . . . . OA08.01 Carpov, A . . . . . . . . . . . P05.06 Carrico, C . . . . . . . . . . OA03.03 Carrington, M . . . . . . . . . P08.03 Carter, D . . . . . . OA06.03, P02.02, P05.03, P19.19 Carville, A . . . . . . OA11.04, P19.27 Casabona, J . . . . . . . . . . P06.06 Casimiro, D . . . . P14.14 LB, S07.01 Casey, E . . . . . . . . . . . . . S07.01 Caskey, M . . . . . . . . . . . S05.04 Cassidy, CM . . . . . . . . . . P10.06 Castro-Orgaz, L . . . . . . . . P09.10 Cavassini, M . . . . . . . . . . P20.01 Center, R . . . . . . .OA06.05, P11.01, P11.02 Cervenka, T . . . . . . . . . P20.07 LB Cesa, K . . . . . . . . . . . . OA06.01 Chaikummao, S . . . .P06.12, P20.04 Chakauya, E . . . . . . . . . . P15.04 Chakrabarti, B . . OA07.03, OA10.05, P19.32 LB Chakrapani, V . . . . . . . . . P16.07 Chalifour, A . . . . . . . . . OA10.03 Champiat, S . . . . . . . . . . P17.21 Chan, B . . . . . . . . . . . . P08.04 Chan, ES . . . . . . . . . . . . P09.01 Chan-Hui . . . . . . . . . . . S02.05 Chang, J . . . P09.01, P09.02, P09.07 Chang, JT . . . . . . . . . . OA02.01 Chapman, R . . . . . . P11.07, P18.11 Chariyalertsak, C . . . . . . . P06.04 Chariyalertsak, S . . . . . . . P06.04 Charuthamrong, P . . . . . . P06.05 Chaussabel, D . . . . . P01.06, S05.02 Chebloune, Y . . . . . . . . . . P19.11

DSouza, P . . . . . . . . . . . P06.03 Dagna, L . . . . . . . . . . P06.15 LB Dai, A . . . . . . . . . . . . OA08.04 Dai, K . . . . . . . . . . . . OA03.03 Dally, L . . . . . . OA09.01, P14.12 LB Dambach, K . . . . . . . . . . P01.05 Daneau, G . . . . . . . . . . . P09.06 Daneau, G . . . . . . . . . P17.30 LB Daniels, R . . . . . . . . . . . P18.17 Daniuk, C . . . . . . . . . . . P18.12 Darrah, PA . . . . . . . . . . . P17.22 Das, A . . . . . . . . . . . P17.28 LB David, WB . . . . . . . . . . OA01.02 Davis, D . . . . . . P04.04, P04.35 LB Davis, I . . . . . . OA05.01, OA05.06, P17.12 De Crignis, E . . . . . . . . . . P20.01 De Baker . . . . . . . . . . . . S03.02 De Groot, A . . . . . P06.07, P18.19, P18.20 De Jager, PL . . . . . . . . . OA06.01 De Oliveira, T . . . . . . . . . P20.05 De Rosa, S . . . . OA10.06, P18.25 LB De Rose, R . . . . . . . . . . OA08.07 De Schacht, C . . . . . . . . . P06.10 De Souza, M . . . . . . . OA01.06 LB, OA05.03, OA07.08 LB, OA09.04, P04.13, P10.07 LB, P17.29 LB, P14.13 LB, P20.04 De Stradis, A . . . . . . . . . . P19.15 Debra, D . . . . . . . . . . . OA06.04 DeCamp, A . . . . . . . OA08.08 LB, OA09.05, P01.08, P18.28 LB, S07.04 DeCastro, C . . . . . . . . . . P06.03 Decker, JM . . . . . . . . . . . P04.07 Decoville, T . . . . . . P04.23, P04.26 Deeks, S . . . . . . . . . . . OA07.05 Deeks, SG . . . . . . . . . . . P17.21 Defawe, OD . . . . . . . . . OA06.03 Defechereux, P . . . . . . . P18.26 LB Del Medico Zajac, MP . . . . . P18.07 Delair, T . . . . . . . . . . . . P19.16 Delaire, T . . . . . . . . . . . P02.05 Deng, W . . . . . . . . . . . . S07.01 Denner, J . . . . . . . . . . . P05.02 Denny, L . . . . . . . . . . . . P10.02

Eamsila, C . . . . OA01.06 LB, P06.05 Earl, P . . . . . . OA01.02, OA01.05, P02.08, P14.05, P19.09, P19.16 Eaton, N . . . . . . . . . . . . P15.11 Eberhard, JM . . . . . . . . . P09.05

232

AIDS Vaccine 2011

Author Index
Edlefsen, PT . . . . P18.28 LB, S07.01, S07.02, S07.04 Edupuganti, S . . . . . . . P18.25 LB Eldridge, J . . . . . . . . . P18.25 LB Eligius, LF . . . . . . . . . . . P14.06 Elizaga, M . . . . OA02.01, P18.25 LB Ellefsen-Lavoie, K . . . . . P18.21 LB Eller, L . . . . . . . OA05.03, P04.13, P14.05 Eller, M . . . . . . . OA05.03, P04.13, P14.05 Else, J . . . . . . . . . . . . OA08.01 Epaulard, O . . . . . . . . . . P17.23 Eriksson, EM . . . . . . . . P18.26 LB Eser, A . . . . . . . . . . . . OA05.03 Eskin, E . . . . . . . . . . . . P18.14 Esparza, J . . . . . . . . . . . P16.04 Essack, Z . . . . . . . . . . . . P16.07 Essex, M . . . . . . .OA05.05, P12.03 Esteban, M . . . . . OA01.01, P14.10, P18.05, P18.07, P19.18, P19.19, P19.21 Estes, J . . . . . . . . . . . . OA08.01 Etemad, B . . . . . . . . . . . P04.20 Euler, Z . . . . P04.06, P04.16, P04.24 Evans, DT . . . . . . OA09.02, P19.27 Evans, TI . . . . . . . . . . . OA11.02 Excler, J . . . . . P14.12 LB, OA09.01, P14.03, P18.06 Flamar, A . . . . . . . . . . OA03.01 Flanagan, K . . . . .OA09.06, P12.02 Flanders, M . . . . OA05.01, OA05.06, P17.12 Fletcher, J . . . . . . . . . . . P01.03 Fleury, S . . . . . . . OA10.03, S02.01 Fling, S . . . . . . . . . . . . . S02.05 Flood, D . . . . . . . . . . . . P06.11 Foley, BT . . . . . . . . . . . . P20.01 Fomsgaard, A . . . . . . . . . P13.01 Fong, Y . . . . . . . . . . . OA06.03 Forsman, A . . . . . . . . . . P04.14 Fouda, G . . . . . . .OA04.03, P12.01 Fraham, N . . . . . . . . . . . P01.08 Frahm, N . . . . . OA06.03, OA10.06, P01.02, P18.27 LB , P18.28 LB, S07.01 Franchini, G . . . OA01.03, OA08.06, OA11.03, P07.02, P19.25 Francis, D . . . . OA01.06 LB, P14.03, P14.13 LB Franco, D . . . . . . . . . . . OA07.01 Freel, S . . . OA06.02, P09.15, P17.16 Freitas, I . . . . . . . . . . . . P04.20 Fras, M . . . . . . . . . . . . P09.10 Friedrich, DP . . . OA10.06, P18.27 LB Fritts, L . . . . . . . . . . . P14.14 LB Fu, H . . . . . . . . . . . . . . P19.28 Fuchs, J . . . . . . . . P06.11, P15.11 Ghobbeh, A . . . . . . . . . OA10.05 Giavedoni, LD . . . . . . . . . P19.26 Gichuhi, C . . . . . . . . . . . P12.02 Gilbert, A . . . . . . . . . . OA02.03 Gilbert, P . . OA01.06 LB, OA08.08 LB, OA09.04, P01.01, P01.08, P14.13 LB, P14.14 LB, P18.14, P18.27 LB, P18.28 LB, S07.04, S07.01 Gill, D . . . . . . OA09.01, P17.29 LB Gillis, J . . . . . . . . OA11.02, P17.10 Gilmour, J . . . . OA09.01, P14.12 LB, P17.29 LB, P20.09 LB Ginsberg, A . . . . . . . . . . P19.24 Giraldo-Vela, J . . . . . . . . OA08.02 Gladden, AD . . . . . . . . . . P08.03 Glidden, DV . . . . . . . . P18.26 LB Gobet, B . . . . . . . . . . . . P16.08 Godoy-Ramirez, K . . . . . . OA09.03, P14.04 Goepfert, P . . . . OA02.01, OA10.06, P08.01, P17.06 Goldstein, H . . . . . . . . . . P14.02 Gmez, C . . . . . . . P14.10, P18.07, P19.18, P19.21 Gonalves, E . . . . . . . . . . P06.10 Gonzlez, R . . . . . . . . . . P09.10 Gonzlez-Sanz, R . . . . . . . P14.10 Gonzalez-Scarano, F . . . . OA08.01 Goodenow, MM . . . . . . . OA05.04 Goodier, S . . . . . . . . . . . P17.20 Goodwin, D . . . . . . . . P18.24 LB Goodwin, M . . . . . . . . P06.14 LB Gordon, ML . . . . . .P20.05, P21.04 Gordon, S . . . . . OA01.03, OA11.03 Gorny, M . . . . . . OA04.02, P04.30, P04.32, P04.41 LB Gorry, P . . . . . . . . P11.01, P11.02 Gottardo, R . . . . . . . . . OA06.03 Goulder, P . . . . . . . . . P07.04 LB Govender, P . . . . . . . . . . P17.01 Goyal, R . . . . . . . . . . P14.12 LB Graham, B . . . . .OA10.06, OA11.03, S04.01 Grant, RM . . . . . . . . . P18.26 LB Grasperge, BF . . . . . . . . . P02.04 Gray, CM . . . . . . .OA06.04, P17.20 Gray, ES . . . . OA04.07 LB, OA07.02, P04.03, P04.07, P04.24, P04.29, P08.02, P15.04 Gray, G . . . . . . . . P09.11, P16.03, P18.28 LB Gray, LR . . . . . . . .P11.02, SO1.02 Graziosi, C . . . . . . . . . . . P20.01 Greene, JM . . . . . . . . . P17.31 LB Greene, KM . . . . . . . . . . P04.22 Grevstad, B . . . . . . . . . . P04.25 Griesbeck, M . . . . . . . . . P09.01 Grijsen, ML . . . . . . . . . . P04.06 Grinsztejn, B . . . . . . . . . . P20.03 Groden, E . . . . . . . . . . . P09.07 Guan, Y . . . . . . OA01.03, OA10.04, P19.25 Guenaga, F . . . . . . . . . . P04.18 Guenaga, JF . . . . . . . . . OA10.05 Guenthner, PC . . . . . . . . . P21.07 Guglietta, S . . . . . . . . . . P20.01 Guimares, ML . . . . . . . . P04.08 Gulua, N . . . . . . . . . . . . P15.03 Gummuluru, S . . . . . . . . . P04.20 Gupta, P . . . . . . . . . . P15.12 LB Gurunathan, S . . . . . OA01.06 LB, P14.03, P14.13 LB, SS01.02 Guwatudde, D . . . . . . . . . P04.13 Gyenes, G . . . . . . . . . P18.24 LB

Fadda, L . . OA02.02, P09.07, P09.08, P17.15, S03.04 Fairhurst, D . . . . . . . . . . P21.01 Fakunle, O . . . . . . . . . . . P15.10 Falivene, J . . P10.01, P18.07, P18.08 Falkowska, E . . . OA04.01, OA04.05, S02.05 Farmerie, WG . . . . . . . . OA05.04 Faruk, NF . . . . . . . . . . P18.24 LB Fast, P . . . . . . OA09.01, P06.16 LB, P06.15 LB, P14.12 LB, P17.29 LB, P20.09 LB Fathi, A . . . . . . . . . . . . P01.04 Fauci, AS . . . . . . . . . . . . P21.08 Favilene, J . . . . . . . P17.02, P17.07 Febo-Vazquez, I . . . . . . . . P14.11 Felber, B . . . . . OA01.05, OA10.04, P19.22, P19.26 Feng, Y . . . . . . . . . . . . OA07.03 Fenizia, C . . . . . OA01.03, OA11.03, P07.02, P19.25 Ferber, M . . . . . . . . . . P07.05 LB Fernandez, C . . . . . . . . OA08.07 Fernndez, E . . . . . . . . . . P06.06 Ferrari, G . . . . . OA01.03, OA06.02, P06.03, P09.15, P12.01, P19.25 Ferrari for the CHAVI and CAVD-CAVIMC Teams . . . . P09.16 Ferreira, CB . . . . . . . . . . . P11.06 Fvrier, M . . . . . . . . . . . P18.15 Fiebig, U . . . . . . . . . . . . P05.02 Fields, SD . . . . . . . . . . . P15.08 Filali, A . . . . . . . . . . . . . S05.04 Filinova, E . . . P03.02 LB, P19.31 LB Fischer, K . . . . . . . . . . . P18.17 Fischetti, L . . . . . . . . . . . P10.03 Fischl, M . . . . . . . . . . . . P05.04 Fisher, KA . . . . . . . . . . . P16.08 Flach, B . . . . . . . . . . . . P04.13

Gabriel, E . . . . . . . . . . . P01.08 Gaekwad, J . . . . . . . . . . P05.06 Gagarina, E . . . . . . . . . P03.02 LB Galkin, A . . . . P03.02 LB, P19.31 LB Gall, J . . . . . . . . . . . . . P19.20 Gallart, T . . . . . . . . . . . . P09.10 Gallo, R . . . . . . . . . . . . S02.02 Gangadhara, S . . . . . . . . P02.08 Ganoza, C . . . . . . . . . . . P06.06 Gao, H . . . . . . . .OA09.02, P04.22 Gao, Y . . . . . . . . . . . P18.22 LB Garced, S . . . . . . . . . . . . P14.11 Garcia, A . . . . . . . . . . . . P06.03 Garca, F . . . . . . . P09.10, P11.04, P11.05, P14.10 Garca-Arriaza, J . . . P14.10, P19.18 Garcia-Morales, C . . . . . P07.04 LB Garca-Perez, JJ . . . . P11.04, P11.05 Garland, MJ . . . . . . . . . . P10.06 Garrido-Rodriguez, D . . . P07.04 LB Garrison, KE . . . . P17.21, P18.24 LB Gartlan, K . . . . . . . P02.09, P02.10 Gaseitsiwe, S . . . . . . . . . P12.03 Gatanaga, H . . . . . . . . . . P17.08 Gatell, JM . . . . . . P06.06, P09.10, P11.04, P11.05, P11.06, P14.10 Gaudieri, S . . . . . . . . . . . P07.01 Geng, Y . . . . . . . . . . . . P19.28 Genin, C . . . . . . . . . . . . P02.05 Georgiev, I . . . P04.37 LB, P04.40 LB Gerstoft, J . . . . . . . . . . . P13.01 Gervassi, A . . . . . . . . . . . P19.23 Gerzmava, O . . . . . . . . . . P15.03 Gettie, A . . . . . . . . . . . . P02.04 Ghebremichael, M . . . . . . P17.12 Gherardi, MM . . . . P10.01, P17.02, P17.07, P18.07, P18.08 Ghiglione, Y . . . . . . P17.02, P17.07

Haase, AT . . . . OA11.01, OA11.06 LB Haberl, A . . . . . . . . . . OA03.04 Habte, H . . . . . . . . . . . OA10.01 Hachaambwa, L . . . . . . . OA09.01 Hahn, B . . . . . . . . . . . . S07.03 Haigwood, NL . . . . . . . . . P05.07 Haining, W . . . . . . . . . . S05.03 Hall, J . . . . . . . . . . . . . P18.15 Hall, O . . . . . . . . . . . . OA01.01 Hamilton, MK . . . . . . . . OA02.01 Hammond, PW . . . . . . . . S02.05 Hammer, S . . . . . . . . . . . P15.11 Han, D . . . . . . . . . . . . OA10.01 Han, X . . . . . . . . . . . . . P04.02 Hance, RJ . . . . . . . . . . P18.26 LB Hanke, T . . . OA09.06, P12.02, P18.01 Hankins, C . . . . . . P16.04, PL02.01 Hannaman, D . . . . . . . P17.29 LB Harari, A . . . . . . .OA01.01, P17.11, P19.20 Harmon, T . . . . . . . . . . . P16.08 Harms, T . . . . . . . . . . . . P09.15 Harris, J . . . . . . . . . . . . P18.17 Harris, L . . . . . . . . . . . OA09.02 Harrison, P . . . . . . . . . . . P16.08 Hartog, K . . . . . . . . . . P19.33 LB Harvey, H . . . . . . . . . . . P04.03 Harvey, JD . . . . . .OA09.02, P19.27 Hasenkrug, A . . . . . . . . . P17.21 Hawkins, NR . . . . . . . . . . P01.01 Hayes, P . . . . . OA09.01, P17.29 LB Haynes, B . . . . . OA03.03, OA04.03, OA06.02, OA09.05, OA09.04, OA10.06, P01.06, P04.24, P04.36 LB, P04.39 LB, P04.40 LB, P09.16, PL01.04, S05.02 Haynes for the CHAVI and CAVD-VIMC Teams . . . . . . P09.15 He, D . . . . . . . . . . P01.01, P18.14 He, X . . . . . . . . . . . . . OA08.05 Heath, S . . . . . . . . . . . . P17.06 Hecht, F . . . . OA05.06, OA07.05, P 17.21 Heckerman, D . . . . P17.06, P17.12, S03.04 Heeney, J . . P04.04, P18.05, P19.33 LB Heipertz, RA . . . . . . . . P20.07 LB Hejdeman, B . . . . .OA09.03, P14.04 Hendry, R . . . . . . . . . . . P21.07 Hensley, TR . . . . . . . . . OA02.01 Herath, S . . . . . P17.33 LB, P18.17, P18.23 LB Herbeck, JT . . . . . . . . . . P01.02 Herman, MM . . . . . . . . . P17.05 Hermanus, T . . . . . . . OA04.07 LB Herrera, E . . . . . . . . . . OA01.02 Hertz, T . . . P01.01, P07.01, P18.14, P18.27 LB, P18.28 LB, S07.01, S07.04 Hervouet, C . . . .P17.33 LB, P18.17, P18.23 LB Heyndrickx, L . . . P04.25, P04.35 LB

AIDS Vaccine 2011

233

AUTHOR INDEX

Author Index
Hidajat, R . . . . . . . . . . OA11.03 Higgins, K . . . . . . .P04.33, P04.34 Hilt, S . . . . . . . . . . . . P19.33 LB Hines, PJ . . . . . . . . . . P17.31 LB Hirsch, V . . . . . OA01.05, OA10.04, P02.08, P07.02 Ho, DD . . . . . . . . OA07.01, S04.02 Ho, J . . . . . . . . . . . . . . P17.22 Hocini, H . . . . . . . . . . . P14.07 Hodara, VL . . . . . . . . . . . P19.26 Hodge, G . . . . . . . . . . P14.14 LB Hoffenberg, S . . . . .P05.06, P19.04 Hogerkorp, C . . . . . . . . OA07.03 Holditch, SJ . . . . . . . . P18.26 LB Holl, V . . . . OA07.04, P04.23, P04.26 Holt, NG . . . . . . . . . . P07.05 LB Holtz, TH . . . . . . . . . . . . P06.12 Honda, M . . . . . . . . . . . P18.04 Hong, CM . . . . . . . . . . OA04.06 Hong, H . . . . . . . .P09.05, P17.10 Hong, K . . . . . . . OA06.06, P04.22 Hong, M . . . . . . . . . . . . P17.21 Honnen, W . . . . . . . . . . P04.30 Hope, T . . . . . . . OA08.03, S06.01 Horton, H . . . . . . P01.01, P18.14, P18.27 LB, P19.23 Hou, J . . . . . . . . . . . . . P02.03 Hou, W . . . . . . . . . . . OA05.04 Howard, W . . . . . . . . . . P17.24 Hoxie, J . . . . . . . . P09.15, P09.16 Hraber, PT . . . . . . . . OA08.08 LB Hryniewicz, A . . . . OA11.03, P19.25 Hu, H . . . . . . . . . P17.17, P17.18 Hu, X . . . . . . . . . . . . . . P04.22 Huang, JC . . . . . . . . . . . P07.01 Huang, X . . . . . . . P17.16, P19.06 Huang, Y . . . . . OA09.02, P14.14 LB Huber, M . . . . . . OA04.05, S02.05 Hue, S . . . . . . . . . . . . . P14.07 Hughes, LJ . . . . . . . . . . . P08.01 Humes, D . . . . . . . . . . . P15.08 Hunter, DV . . . . . . . . . P18.24 LB Hunter, E . . . . . P06.14 LB, P08.01 Hunter, M . . . . . P15.12 LB, P19.24 Hural, J . . . . P18.28 LB, P19.30 LB, S07.01 Huret, C . . . . . . . . . . . . P19.05 Hust, M . . . . . . . . . . . OA03.04 Hutnick, N . . . . . . .P05.08, P19.24 Huynh, N . . . . . . . . . . OA04.04 Hwang, K . . . . P04.36 LB, P04.39 LB James, I . . . . . . . . . . . . P07.01 Janes, H . . . . . . .OA06.03, P01.08 Jani, I . . . . . . P06.09, P06.10, P19.08 Jansson, M . . . . . . . . . . P04.25 Jaoko, W . . . . . . .OA09.06, P12.02 Jaspan, H . . . . . . . . . . . OA11.05 Jean, L . . . . . . . . . . . . . P12.03 Jeffs, S . . . . . . . . . . . . . P21.01 Jegaskanda, S . . . . . . . . OA08.07 Jelicic, K . . . . . . . . . . . . P21.08 Jennes, W . . . . . . . . . . . P09.06 Jernberg, L . . . . . . . . . . . P14.04 Jessen, H . . . . . . OA05.01, OA05.06 Jia, M . . . . . . . OA06.06, OA08.05 Jiang, W . . . . . . . . . . . . P19.06 Jiang, X . . . . . . . . . . . OA10.02 Jimnez, J . . . . . . . . . . . P14.10 Jimnez, V . . . . . . . . . . . P14.10 Jin, X . . . . . . . . . .P04.19, P18.02 Joachim, A . . . . . . . . . . . P14.06 Jobe, O . . . . . . . OA02.05, P02.06 John, M . . . . P07.01, P08.04, S03.01 John, VM . . . . . . . . . . P18.24 LB John-Stewart, G . . OA09.06, P12.02 Johnson, J . . . . . . . . . P10.08 LB Johnson, P . . . . . . . . .OA11.06 LB Johnson, R . . . . . .OA11.02, P03.01, P17.10, P19.27 Johnson, RP . . . . . . . . . OA08.03 Johnson, S . . . . . . . . . . OA08.06 Johnson, W . . . . . . . . . . S03.05 Jojic, N . . . . . . . . . . . . . P07.01 Jones, BR . . . . . . . . . . P18.24 LB Jongrakthaitae, S . . . . . P10.07 LB Jonsdottir, L . . . . . . . . . . P15.09 Jose, HE . . . . . . . . . . . . P12.03 Joseph, S . . . . . . . . . . . P19.19 Jost, S . . . . . . . . . . . . . P09.13 Jourdain, G . . . . . . . . . . P21.03 Joyce, M . . . . . . . . . . P04.37 LB Juarez-Molina, C . . . . . . P07.04 LB Julien, J . . . . . . . OA04.05, S02.05 Jurgens, C . . . . . . . . . . . P19.04 P08.01, P20.09 LB Karl, JA . . . . . . . . . . . OA08.04 Karln, K . . . . . . .OA09.05, P14.04 Karlsson, AC . . . . . . . . . . P17.09 Karlsson, I . . . . . . .P13.01, P17.19 Karlsson Hedestam, GB . . . OA04.04 Karuna, S . . . . . . . . . . . . P15.11 Kasprowicz, VO . . . . . . . . P17.01 Katende, D . . . . . . . . . . . P15.07 Kathuria, N . . . . . . . . . . P19.24 Katlama, C . . . . . . . . . . . P19.08 Katze, MG . . . . . . . . . . OA08.04 Katzenstein, TL . . . . . . . . P17.19 Kaufmann, DE . . . . OA06.01, P17.01 Kaulitz, D . . . . . . . . . . . P05.02 Keefer, M . . . . . OA09.01, OA10.06, P04.19, P06.08, P15.08, P18.02 Keele, B . . . . . . OA01.03, OA10.04, OA11.03, P07.02, P19.25, S07.05 Kelleher, A . . . . . OA05.06, P04.05, P11.01, P17.32 LB Kemper, M . . . . . .OA03.02, P08.03 Kennedy, E . . . . . . . . . . . P18.02 Kent, SJ . . . . . . OA02.04, OA06.05, OA08.07, P04.05, P17.32 LB Kersh, EN . . . . . . . . . . . P21.07 Kestens, L . . . . . P09.06, P17.30 LB, P21.05 Keynan, Y . . . . . . . . . . . P09.09 Khan, A . . . . . . OA01.02, OA08.04, P05.08, P18.16, P18.25 LB, P19.24 Khan, A . . . . . . . . . . . . P19.01 Khatamzas, E . . . . . . . . . P09.14 Kiazyk, S . . . . . . . .P17.03, P17.05 Kibengo, FM . . . . . . . . . . P15.07 Kidega, W . . . . . . . . . . . P16.06 Kijak, G . . . . . . . . . . . OA07.06 Kilembe, W . . . P06.14 LB, P20.09 LB Kim, B . . . . . . . . . . . . . P18.02 Kim, J . . . . . . . OA01.02, OA01.03, OA01.06 LB, OA05.03, OA06.03, OA07.08 LB, OA09.02, OA09.05, OA09.04, OA10.06, P01.03, P01.05, P01.07, P10.07 LB, P14.03, P14.05, P14.13 LB, P18.06, P20.07 LB, P20.08 LB, S07.01 Kim, M . . . . . . . . . . . . . S07.01 Kimani, J . . . . . . . P09.09, P17.03, P17.04, P17.05, P18.12, PL01.03 Kimani, M . . .P17.03, P17.04, P18.12 Kines, RC . . . . . . . . . . . OA11.03 King, B . . . . . . . . . . . . OA03.01 King, CR . . . . . . . . . . . PL03.02 King, D . . . . . . . . . P02.02, P10.03 King, M . . . . . . . . . . . . P01.04 King, R . . . . . . . . . . . . . P05.06 Kiravu, A . . . . . . . . . . . . P11.07 Kirchherr, J . . . . . . . . . OA06.02 Klaric, K . . . . . . . . . . . OA04.03 Klatt, NR . . . . . . .OA08.01, P17.22 Klatzmann, D . . . . . . . . . P19.05 Klavinskis, L . . . .P17.33 LB, P18.17, P18.23 LB Kliche, A . . . . . . . . . . . . P04.10 Kobie, JJ . . . . . . . . . . . . P04.19 Kobinger, G . . . . . . . . . . P18.10 Koblin, B . . . . . . . . P08.04, P15.11 Koch, S . . . . . . . . . . . OA03.04 Kochar, N . . . . . . . . . . OA06.03 Kochhar, S . . . . . . . . . P14.12 LB Koen, J . . . . . . . . . . . . . P16.07 Koester Kiazyk, SA . . . . . . P17.04 Koff, W . . . . . . . OA04.05, P04.27, P04.42 LB, P04.40 LB, P05.06, S02.05 Koita, O . . . . . . . . P06.07, P18.19, P18.20 Komaroff, W . . . . . . . . OA09.01 Kone, Y . . . . . . . . P06.07, P18.19 Kong, L . . . . . . . . . . . OA04.05 Kong, X . . . . . . OA09.05, OA10.02, P04.41 LB Kongpechsatit, O . . . . . . . P20.04 Konopa, P . . . . . P18.28 LB, S07.01 Koopman, G . . . . . . . . . . P18.05 Koornstra, W . . . . . . . . . P04.04 Kopycinski, J . . . . . . . . . OA09.01, P17.29 LB Korber, B . . . . OA07.02, OA08.08 LB Kosachunhanan, N . . . . . . P06.04 Koty, Z . . . . . . . . P06.07, P18.19, P18.20 Koup, R . . . . . . . . . . . . P06.03 Koutsoukos, M . . . . . . . . P18.15 Kowawisatsut, L . . . . . OA02.06 LB Kozink, D . . . . . . . . . . . P09.15 Kozlov, A . . . . . . . . . . . . P14.09 Kozlowski, P . . . . OA01.05, P02.08, P10.05 Krachmarov, C . . . . . . . . . P04.30 Kraft, Z . . . . . . . . . . . . P05.07 Kramski, M . . . . . . . . . OA06.05 Krasnoselskih, T . . . . . . . . P14.09 Krause, K . . . . . . . . . . P10.08 LB Krawchenko, A . . . . . . . . P18.10 Kraynyak, K . . . . . . . . . . P19.24 Krebs, M . . . . . . . . . . P06.14 LB Kremer, EJ . . . . . . OA01.01, P19.20 Krishnamurty, AT . . . . . . OA02.01 Kronborg, G . . . . . . . . . . P13.01 Kuang, T . . . . . . . . . . . . P08.04 Kublin, J . . . . . . . . . . P18.28 LB Kuebler, PJ . . . . . . . . . P18.26 LB Kuhn, L . . . . . . . . . . . . . P09.11 Kulkarni, V . . . . . .OA10.04, P19.22 Kumar, S . . . . . . . . . . . OA02.02 Kunasol, P . . OA01.06 LB, P14.13 LB Kunwar, P . . . . . . . P01.01, P18.14 Kurle, S . . . . . . . . . . . P14.12 LB Kuroiwa, J . . . . . . . . . P20.07 LB Kurth, R . . . . . . . . . . . . P05.02 Kutsyna, G . . . . . . . . . . OA11.03 Kutzler, M . . . . . . . . . . . P19.24 Kuwata, T . . . . . . . . . . . P04.21 Kuzmichev, Y . . . . OA11.02, P03.01 Kuznecovs, G . . . . . . . P03.02 LB, P19.31 LB Kwa, S . . . . . . . . OA01.05, P02.08 Kwissa, M. . . . . . . . . . . . S05.01 Kwon, D . . . . . . . . . . . OA05.06 Kwon, S . . . . . . . . . . . . P18.17 Kwong, P . . . . . OA03.03, OA04.01, OA04.03, P04.14, P04.36 LB, P04.37 LB, P04.39 LB, P04.42 LB, P04.40 LB, P05.06, PL03.01, S02.04

AUTHOR INDEX

Ibrahim, S . . . . . . .P20.05, P21.04 Igarashi, T . . . . . . . . . . . P04.21 Inambao, M . . . . . . . . . . P04.09 Ingabire, R . . . P06.15 LB, P06.16 LB Ip, S . . . . . . . . . . . . . P17.32 LB Isitman, G . . . . OA02.04, OA06.05, P04.05 Ismael, N . . . . . . . . . . . P06.10 Issa, K . . . . . . . . . . . . . P15.10 Iyer, S . . . . . . . . . . . . . . S07.01

Jackson, D . . . . . . . . . . . P11.01 Jacob, RA . . . . . . . . . . . P04.28 Jacobs, B . . . . . . . . . . . . P18.05 Jkel, M . . . . . . . . . . . . P09.05 Jalah, R . . . . . . . . . . . OA10.04

Kabra, S . . . . . . . . . . P20.06 LB Kaeratiswetanun, W . . . . . P06.05 Kaewkungkal, J . . . . . OA01.06 LB, OA07.08 LB, OA09.02, P14.03, P14.13 LB Kaewkunwal, J . . . . . . . OA09.04 Kagee, A . . . . . . . . . . . . P16.03 Kalams, S . . . . . P05.07, P18.25 LB Kaleebu, P . . . . . . P15.05, PL01.01 Kalil, J . . . . . . . . .P18.13, P18.18 Kalls, EG . . . . . . . P17.21, P17.25, P18.18, P18.26 LB Kalra, R . . . . . . P04.17, P20.06 LB Kamali, A . . . . . . . P04.09, P15.05, P15.07, P20.09 LB Kanekiyo, M . . . . . . . . . . P18.04 Kang, H . . . . . . . . . . . . P12.01 Kang, Y . . . . . . . . P15.06, P19.12 Kannanganat, S . . . . . . . . P02.08 Kapiga, SH . . . . . . . . . P04.40 LB Kappes, J . . . . . OA06.02, OA07.06, P09.15, P09.16 Karasavvas, N . OA07.08 LB, OA09.04 Karia, E . . . . . . . . . . . . P04.09 Karim, S . . . . . . . . . . P04.42 LB Karita, E . . . . P06.15 LB, P06.16 LB,

234

AIDS Vaccine 2011

Author Index
La, D . . . . . . . . . . . . . . P18.12 Labranche, C . . . . . . . . OA10.01 Lacabaratz, C . . . . . . . . . P14.07 Lacas, A . . . . . . . . . . . . P09.02 Lacerda, M . . . OA07.02, OA08.08 LB Lai, J . . . . . . . . . . . . . OA04.05 Lai, L . . . . . . . . . OA01.05, P02.08 Lai, Z . . . . . . . . . . . . . . P04.30 Lakhi, S . . . . . . P04.09, P20.09 LB Lallemant, M . . . . . . . . . P21.03 Lam, A . . . . . . . . . . . P18.28 LB Landais, E . . . . . . . . . . . P04.09 Lande, R . . . . . . . . . . . . P16.08 Lane, J . . . . . . . . . . . . . P06.03 Lane, K . . . . . . OA02.02, OA05.01, OA05.06, P09.07, P09.08 Langhammer, S . . . . . . . . P05.02 Lanxon-Cookson, E . . . . . . P19.22 Lanzavecchia, A . . . . . . P04.35 LB Larsen, BB . . . . . . . . . P18.28 LB, S07.01, S07.02 Latka, M . . . . . . . . . . P20.09 LB Lau, C . . . . . . . . OA01.02, P15.06 Lauer, WA . . . . . . . . . . . P19.27 Laufer, D . . . . . . . . . . P06.16 LB Laufer, N . . . . . . . . P17.02, P17.07 Laumond, G . . . . OA07.04, P04.23, P04.26 Launay, O . . . . . . . . . . . P14.01 Lawson, B . . . . . . . . . . OA08.01 Lazzarin, A . . . . . . . . . P18.21 LB Le, AQ . . . . . . . . . . . . . P08.04 Le Gall, S . . . . . . . . . . . P19.22 Le Grand, R . . . . P10.10 LB, P14.08, P17.23, P19.16 Le Heron, A . . . . . . . . . . P18.17 Lederle, A . . . . . . OA07.04, P04.23 Lee, J . . . . . . . . . . . . . OA01.02 Lee, W . . . . . . . . . . . . . P16.08 Leelawiwat, W . . . . . . . . . P20.04 Lefebvre, F . . . . . . . . . . . S05.04 Legg, H . . . . . . . . . . . P19.33 LB Lehman, J . . . . . . . . . . . P17.21 Lei, E . . . . . . . . . . . . P20.08 LB Lelivre, J . . . . . . . P14.01, P14.07 Lemoine, B . . . . . . P01.06, S05.02 Leon, A . . . . . . . . . . . . P06.06 Leon, EJ . . . . . . . . . . . OA08.02 Lepelley, A . . . . . . . . . . . P09.12 Leroux-Roels, G . . . . . . . OA10.03 Lertmemongkolchai, G . . . . P01.06, S05.02 Leslie, AJ . . . . . . . . . . . . P09.14 Letvin, NL . . . . . . . . OA08.08 LB Levitz, L . . . . P06.07, P18.19, P18.20 Lvy, Y . . . . . . OA01.01, OA02.03, OA03.01, P01.06, P10.10 LB, P14.07, P14.01, P14.08, S05.02 Lewis, G . . . . . . . . . . . . S02.02 Lewis, MG . . . . . . . . . . OA08.04 Lhost, JJ . . . . . . . . . . P17.31 LB Li, B . . . . . . . . . OA08.01, P18.15 Li, D . . . . . . . . OA06.06, OA08.05 Li, F . . . . . . . . . . . . . . P18.17 Li, F . . . . . . . . . . . . . P18.23 LB Li, H . . . . . . . OA11.04, P10.08 LB Li, J . . . . . . . . . OA02.01, P04.01 Li, Q . . . . . . OA11.01, OA11.06 LB Li, S . . . . . . OA01.06 LB, P14.13 LB

Li, W . . . . OA08.03, P15.06, P19.27 Li, Y . . . . . . . . OA04.04, OA07.03, OA08.05 Liang, H . . . . . .OA06.06, OA08.05 Liao, H . . . . . . OA04.03, OA10.06, P04.36 LB, P04.39 LB Liebenberg, L . . . . . . . . . OA11.05 Liebl, B . . . . . . . . . . . . . P06.03 Lifson, JD . . . . . OA01.03, OA08.06, OA10.04, OA11.06 LB, P09.07, P17.22, P19.25, P19.27 Limoli, K . . . . . . . .P04.33, P04.34 Lindegger, G . . . . . . . . . . P16.07 Lindqvist, M . . . OA05.01, OA07.05, P17.12 Lindsay, R . . . . . . P09.07, P09.01, P09.02 Ling, C . . . . . . . . P17.26, P19.10, P21.02 Ling, D . . . . . . . . . . . . . P18.09 Linqi, Z . . . . . . . . .P17.26, P19.10 Lioznov, D . . . . . . . . . . . P14.09 Liqiang, F . . . . . . . P19.10, P21.02 Liquet, B . . . . . . . . . . . . P14.07 Little, S . . . . . . . . . . . OA05.06 Liu, B . . . . . . . . . . . . . P19.28 Liu, F . . . . . . . . . . . . P10.09 LB Liu, G . . . . . . . . . . . . P07.03 LB Liu, H . . . . . . . . . . . . . P19.12 Liu, J . . . . . . . OA11.04, P10.08 LB Liu, L . . . . OA05.04, P15.06, P19.12 Liu, M . . . . . . . . . . . . . P18.09 Liu, S . . . . . . . . . . . . P17.32 LB Liu, Y . . . . P01.01, P10.09 LB, P18.09 Liu, Z . . . . . . . . . . . . . . P18.09 Lchelt, M . . . . . . . . . . . P05.02 Lochoshvili, A . . . . . . . . . P15.03 Lodha, R . . . . . . . . . . P20.06 LB Logan, K . . . . . . . . . . . . P18.17 Logan, M . . . . . . . . . . P18.28 LB Logie, C . . . . . . . . . . . . P16.07 Lohman-Payne, B . . . OA09.06, P12.02 Lombardi, K . . . . . . . . . OA07.06 Lombardo, A . . . . . . . . . OA09.01 Longo, N . . . . . . . . . . . OA07.03 Lopalco, L . . . . . . . . . . OA10.03, P11.03 Lpez Bernaldo de Quirs, J . . P14.10 Lorenz, I . . . . . . . . . . . . P19.04 Lorin, CM . . . . . . . . . . . P18.15 Louder, M . . . . . OA04.03, P04.24, P04.27, PL03.01 Louder, RK . . . . . . . . . P04.36 LB Loughran, K . . . . . . . . . OA09.01 Louzao, R . . . . . . . . . . . P06.03 Lowe, AC . . . . . . . . . . OA05.04 Lowy, DR . . . . . . . . . . . OA11.03 Lu, NJ . . . . . . . . . . . . P04.37 LB Lu, S . . . . . . . . . OA10.02, P04.31 Lu, X . . . . . . . . . .P15.06, P19.12 Luallen, RJ . . . . . . . . . . . P19.28 Luedemann, C . . . . . . OA08.08 LB Lund, O . . . . . . . . . . . . P17.09 Lundegaard, C . . . . . . . . . P17.09 Luo, M . . . . . . . . P17.03, P18.10, P18.12 Luo, Z . . . . . . . . . . . . OA08.05 Luster, AD . . . . . . . . . . . P17.15 Luthra, K . . . . . . P04.17, P20.06 LB Lynch, D . . . . . . . . . . . OA11.04 Lynch, R . . . . . . . .P04.07, P04.24

M, M . . . . . . . . . . . . P14.12 LB Ma, Z . . . . . . . . OA11.03, P07.02, P14.14 LB, P19.25 Ma, P . . . . . . . . . . . . OA06.06 Ma, Y . . . . . . . . . . . . . P18.09 MacCormack, S . . . . . . . . P19.08 Macovela, E . . . . . . . . . . P06.10 Madenwald, T . . . . . . . . . P15.11 Madiga, MC . . . . . . . . . . P04.03 Madnote, S . . . . . . . . OA07.08 LB Maekanantawat, W . . . . . . P14.03 Maes, C . . . . . . . . . . . OA10.03 Maesela, P . . . . . . . . . . . P16.03 Maeto, C . . . P10.01, P18.07, P18.08 Magaret, C . . . . P18.28 LB, S07.01, S07.04 Mahan, A . . . . . . . . . . . . P04.11 Mahlokozera, T . . . . . . . . P12.01 Maillard, M . . . . . . . . . OA01.01 Mailliard, RB . . . . . . . . . P19.06 Mairena, EC . . . . . . . . . . P18.18 Maisaka, M . . . . . . . . . P20.07 LB Makhema, J . . . . . . . . . . P12.03 Malcolm, R . . . . . . . . . . P10.06 Malherbe, DC . . . . . . . . . P05.07 Malia, J . . . . . . . . . . . P20.07 LB Mallal, S . . . . . . . . P07.01, P08.04 Maness, NJ . . . . . . . . . . P17.21 Mangeot, I . . . . . . . . . P10.10 LB Mangeot-Mderl, I . . . . . P14.08 Manhas, S . . . . . . . . . . . P04.32 Mann, JF . . . . . . . . . . . . P19.14 Mann, T . . . . . . . . . . . . P17.06 Manocheewa, S . . . . . . . . P19.22 Manoussaka, M . . . . . . P17.28 LB Mansfield, KG . . . . OA11.04, P19.27 Marais, J . . . . . . . . . . P18.28 LB Marin, E . . . . . . . . . . . . P04.19 Markle, T . . . . . . . . . . . . P08.04 Markowitz, M . . . . . . . . . P08.04 Marone, R . . . . . . . . . . . P15.01 Marone, R . . . . . . . . . . . P15.02 Marovich, M . . . OA01.02, OA05.03, OA07.06, OA09.05, P01.03, P01.05, P14.04, P14.05, P14.06, P18.06 Martha, N . . . . . . . . . . . P16.01 Martin, E . . . . . P08.04, P18.24 LB Martin, JN . . . . . . . . . . . P17.21 Martin, L . . . . . . . . . OA03.05 LB Martin, R . . . . . . OA01.01, P19.20 Martin, W . . . . . . .P18.19, P18.20 Martinez-Guzman, D . . . P19.33 LB Martinon, F . . . . . . . . P10.10 LB, P14.08, P17.23 Marx, P . . . . . . P15.12 LB, P19.24 Mascola, J . . . . OA03.03, OA04.01, OA04.03, OA04.04, OA07.03, OA08.08 LB, P04.07, P04.24, P04.27, P04.36 LB, P04.37 LB, P04.39 LB, P04.42 LB, P04.40 LB, P05.06, P19.32 LB, PL03.01 Matsuo, K . . . . . . . . . . . P18.04 Matsushita, S . . . . . . . . . P04.21 Mattiaco, J . . . . . . . . . . P04.19 Matyas, GR . . . . . OA02.05, P02.07, P21.06

Maust, BS . . . . . . . . . . . S07.01 Mayer, A . . . . . . . . . . . . . . . . P09.14 Mayer, K . . . . . . . . . . . . P08.04 Mbewe, A . . . . . . . . . . . P18.01 Mboup, S . . . . . P09.06, P17.30 LB, P21.05 McChesney, M . . . . . . . P14.14 LB McCluskey, M . . . . . . . . . P16.04 McConnell, JJ . . . . . . . P18.26 LB McCoy, L . . . . . . . . . . . . P04.14 McElrath, MJ . . . OA02.01, OA06.03, OA10.06, P01.08, P18.27 LB, P18.28 LB, S07.01 McKay, P . . . P02.02, P10.06, P19.14, P19.19 McKee, K . . . . . P04.27, P19.32 LB McKinnon, LR . . . . .P17.04, P17.05 McLellan, J . . . OA03.03, P04.36 LB Mclinden, R . . . . . . . . P20.08 LB McMahon, J . . . . . . . . . . P15.04 McMahon, V . . . . . . . . P18.26 LB McManus, J . . . . . . . . . . P02.04 McMichael, A . . . OA06.02, P01.06, P09.14, P17.16, S05.02 McMichael, S . . . . . . . . . P19.07 McMullen, A . . . . . . . . . OA06.01 McNicholl, J . . . . . P06.12, P20.04, P21.07 McRaven, M . . . . . . . . . OA08.03 Mee, E . . . . . . . . . . . . . P18.15 Meguid, N . . . . . . . . . . . P09.16 Mehendale, SM . . . . . . P14.12 LB Mehrotra, M . . . . . . . . P18.26 LB Meiser, A . . . . . P17.33 LB, P18.17, P18.23 LB Mejias, A . . . . . . . . P01.06, S05.02 Melhem, NM . . . . . . . . . P19.06 Melief, C . . . . . . . . . . . . P18.05 Melief, CJ . . . . . . . . . . . P18.01 Mellors, E . . . . . . OA03.02, P08.03 Mendoza, D . . . . . . . . . OA08.06 Mendoza, J . . . . . . . . . . P19.01 Mendoza, M . . . . . . . . . . P17.03 Menezes, A . . . . . . . . . . S01.04 Mens, H . . . . . . . . . . . . P17.19 Mercier, P . . . . . . . . . . . P19.16 Merino-Mansilla, A . . . . . . P11.06 Mestecky, J . . . . . . . . . . P19.24 Metch, B . . . . . . P06.11, P18.25 LB Meulbroek, M . . . . . . . . . P06.06 Meyer, T . . . . . . . . . . . OA03.04 Meyer-Olson, D . . . . . . . . P09.05 Meyers, A . . . . . . . P09.09, P11.07 Mhalu, F . . . . . . . . . . . . P14.06 Michael, N . . . . OA01.02, OA01.03, OA01.06 LB, OA05.03, OA06.03, OA07.06, OA07.08 LB, OA09.02, OA09.05, OA09.04, P01.05, P01.07, P04.13, P10.07 LB, P14.03, P14.05, P14.06, P14.13 LB, P16.04, P20.07 LB, P20.08 LB, S07.01 Michelin, O . . . . . . . . . P07.05 LB Migalska, K . . . . . . . . . . P10.06 Miguel Benito, J . . . . . . . OA03.04 Migueles, SA . . . . . . . . OA08.06 Mihajlovic, V . . . . . . . . P18.24 LB

AIDS Vaccine 2011

235

AUTHOR INDEX

Author Index
Miiro, J . . . . . . . . . . . . P15.05 Miller, CJ . . . . . . OA11.03, P07.02, P14.14 LB, P19.25 Miller, J . . . . . . . . . . . . P17.21 Milner, D . . . . . . . . . . P10.08 LB Milsom, P . . . . . . . . . . . P15.09 Minevich, G . . . . . . . . . . P09.11 Misher, L . . . . . . . . . . . . P05.07 Mitcham, JL . . . . . . . . . . S02.05 Mkhwanazi, Np . . . . . . . OA01.04 Mlisana, K . . . . . OA06.04, P04.03, P17.20 Mlotshwa, M . . . . OA06.04, P17.20 Mmalane, M . . . . . . . . . . P12.03 Mncube, Z . . . . . . . . . . OA05.02 Mock, PA . . . . . . . . . . . P06.12 Mofenson, L . . . . . . . . . PL02.03 Moir, SL . . . . . . . . . . . . P17.22 Moise, L . . . . . . . . . . . . P18.20 Moldoveanu, Z . . . . . . . . P19.24 Montaner, LJ . . . . . . . . . . P14.11 Montano, SM . . . . . . . . . P15.01 Montefiori, D . . OA01.03, OA04.03, OA07.02, OA07.06, OA09.02, OA10.01, OA10.04, P04.13, P04.22, P04.24, P04.39 LB, P09.15, P09.16, P12.01, P19.25, P19.33 LB, P20.08 LB, P21.01 Montes, A . . . . . . . . . . OA03.01 Montes, M . . . . . . . . . . OA03.01 Montifiore, D . . . . . . . . . P19.24 Moody, MA . . . OA04.03, P04.36 LB, P04.39 LB, P09.15 Moog, C . . . OA07.04, P04.23, P04.26 Mooij, P . . . . . . . . . . . . P18.05 Moore, P . . . . OA04.07 LB, OA07.02, P04.03, P04.42 LB, P08.02, P15.04 Moore, PL . . . . . . . . . . . P04.29 Moquin, S . . . P04.37 LB, P04.40 LB Mora, J . . . . . . . . . . . . S06.03 Moreno-Nieves, UY . . . . . . P09.13 Morgado, MG . . . . .P04.08, P20.03 Morgan, CA . . . . . . . . . OA02.01 Morgan, P . . . . . . P01.07, P06.05, P14.03 Morgan, T . . . . . . . . . . . P19.25 Mori, M . . . . . . . . . . . . P05.07 Morineau-Le Houssine, P . . . P14.01 Morozov, V . . . . . . . . . . P05.02 Morris, D . . . . . . . . . . OA09.02 Morris, L . . . . OA04.07 LB, OA07.02, P04.03, P04.07, P04.24, P04.29, P04.39 LB, P04.42 LB, P08.02, P09.16, P15.04 Moscoso, CG . . . . . . . OA03.05 LB Moseri, A . . . . . . . . . . . P04.15 Moss, B . . . . . . OA01.02, OA01.05, P02.08, P14.05, P19.09 Moss, J . . . . . . . . . . . P15.12 LB Mothe, B . . . . . . . P17.24, P19.22 Mous, K . . . . . . . . . . . . P09.06 Moussa, M . . . . . . . . . . . P19.11 Moyle, M . . . . . . . . . . . S02.05 Moyo, S . . . . . . . . . . . . P12.03 Mpendo, J . . . . . . . . . . . P15.05 Mucavela, E . . . . . . . . . . P06.09 Muchenje, W . . . . . . . . . P16.08 Mueanpai, F . . . . . . . . . . P20.04 Mueller, J . . . . . . . . . . OA09.06 Mueller, M . . . . . . . . . . OA07.05 Mufhandu, H . . . . . . . . . P15.04 Mhle, M . . . . . . . . . . . P05.02 Mujib, S . . . . . . . . . . P18.24 LB Mukamuyango, J . . . . . P06.15 LB, P06.16 LB Mukamwezi, J . . . . . . . P06.15 LB Mulenga, J . . . . . . . . . P04.41 LB Mller-Trutwin, M . . . . . . P09.12 Mullins, J . . . . . . . P01.02, P01.08, P18.28 LB, P19.06, P19.22, S07.01 Mundia, M . . . . . . . . . . . P12.02 Munier, CM . . . . . . . . P17.32 LB Munier, S . . . . . . . . . . . P19.16 Muoz-Fernndez, M . . . . . P14.10 Munseri, P . . . . . . . . . . . P14.06 Murashev, B . . . . . . . . . . P14.09 Mureithi, M . . . . . . . . . OA01.04 Murphy, MK . . . . . . . . . . P08.01 Murtaugh, W . . . . . . . . P20.08 LB Murthy, K . . . . . . . . . . OA03.02 Musonda, R . . . . . . . . . . P12.03 Muthumani, K . . . . .P05.08, P19.01 Myles, DF . . . . . . . . . . . P05.08 NIH/NIAID HIV Vaccine Trial Network (HVTN) . . . P18.27 LB NIH/NIAID HIV Vaccine Trial Collaborative Network . . . PL02.02 Nilsson, C . . . . . . OA09.05, P06.09, P06.10, P14.04, P14.06 Nitayapan, S . . . . . . . . . . P14.03 Nitayaphan, S . . . . . . OA01.06 LB, OA05.03, OA07.08 LB, OA09.02, OA09.04, P01.05, P06.05, P14.13 LB Nixon, DF . . . . . P17.21, P18.24 LB, P18.26 LB Njoku, OS . . . . . . . . . P20.07 LB Njuguna, I . . . . . . . . . . . P12.02 Nkolola, J . . . . . . . . . . . P05.03 Noel, SE . . . . . . . . . . P04.40 LB Nogueron, A . . . . . . . . . . P10.05 Nolan, D . . . . . . . . . . . . P07.01 Nonyane, M . . . . . . . . . OA07.02 Norakidze, I . . . . . . . . . . P15.03 Norante, FA . . . . . . . . . OA08.02 Norstrm, M . . . . . . . . . . P17.09 Novitsky, V . . . . . OA05.05, P12.03 Ntale, R . . . . . . . OA06.04, P04.03 Nyombayire, J . . . . . . . P06.15 LB Nyombayire Mutagisha, J . P06.16 LB P, S . . . . . . . . . . . . . P14.12 LB Pace, CS . . . . . . . . . . . OA07.01 Padte, NN . . . . . . . . . . . P02.04 Page, M . . . . . . . . P18.15, P18.17 Pahar, B . . . . . . . . . . . . P19.24 Pahwa, R . . . . . . . . . . . . P05.04 Pahwa, S . . . . . . . . . . . . P05.04 Paiardini, M . . . . . . . . . OA08.01 Palermo, RE . . . . . . . . . OA08.04 Pallikkuth, S . . . . . . . . . . P05.04 Palucka, K . . . . . . . P01.06, S05.02 Pambuccian, S . . . . . . .OA11.06 LB Pan, R . . . . . . . . . . . P04.41 LB Pancera, M . . . OA03.03, P04.36 LB, P04.37 LB, P04.42 LB, P05.06 Pando, M . . . . . . . P15.01, P15.02 Pankhong, P . . . . . . . . . . P18.16 Pankla, R . . . . . . . P01.06, S05.02 Pantaleo, G . . . . . OA01.01, P17.11, P18.05, P18.21 LB, PL01.05 P19.20, P20.01 Pantophlet, R . . . . .P04.32, P19.29 Papagatsias, T . . . . . . . . . P18.17 Paranjape, R . . . . . . . . P14.12 LB Paris, RM . . . . . . . . OA01.06 LB, P14.13 LB Park, H . . . . . . . . . . . . . P10.05 Parker, J . . . . . . . . . . . . P04.25 Parks, C . . . . . . . . . . . . P19.04 Parniak, MA . . . . . . . . P15.12 LB Parodi, LM . . . . . . . . . . . P19.26 Parsons, MS . . . . . . . . . OA02.04 Pascual, V . . . . . . . P01.06, S05.02 Pascuccio, M . . . . . . . . . P21.08 Pascutti, M . . . . . . . . . . P18.08 Pascutti, MF . . . . . . P10.01, P18.07 Pasmore, J . . . . . . . . . . . P10.02 Passmore, J . . . . . . . . . . OA11.05 Patel, N . . . . . . . . . . . . P21.08 Patel, RK . . . . . . . . . . . . P19.14 Patel, V . . . . . . . . . . . OA10.04 Pattanapanyasat, K . . . OA02.06 LB, P17.27 Pattani, A . . . . . . . . . . . P10.06 Patterson, N . . . . . . . . . . P02.04 Patterson, S . . . .P17.33 LB, P18.17, P18.23 LB Pau, M . . . . . . . . . . . P10.08 LB Paul, S . . . . . . . . .P02.05, P19.16 Pavlakis, G . . . . OA01.05, OA10.04, P19.22, P19.26 Pavot, V . . . . . . . . . . . . P02.05 Paximadis, M . . . . . P09.03, P09.11 Peachman, KK . . . .OA02.05, P02.06 Peel, S . . . . . . OA05.03, P20.07 LB Pegu, P . . . . . . . OA01.03, P07.02 Pei-Jen Lai, R . . . . . . . . P19.33 LB Pejchal, R . . . . . . OA04.05, S02.05 Pea, J . . . . . . . . . . . . . P09.10 Peng, H . . . . . . . OA06.06, P18.09 Penichon, J . . . . . . . . . . P04.23 Penney, K . . . . . . . . . . . P08.04 Perdiguero, B . . . . . P14.10, P18.07, P19.21 Peressin, M . . . . . . . . . OA07.04 Pereyra, F . . . . OA02.02, P07.05 LB, P08.03, P09.02, P09.08, P17.12

AUTHOR INDEX

Nabel, G . . . . OA03.03, OA08.08 LB, P04.24, P04.37 LB, P04.39 LB, PL03.01 Nadas, A . . . . . . . . . . . OA10.02 Naider, F . . . . . . . . . . . . P04.15 Nair, K . . . . . . . . . . . . OA05.02 Najburg, V . . . . . . . . . . . P09.12 Njera, J . . . P14.10, P19.18, P19.21 Nakaya, HI . . . . . . . . . . . S05.01 Nakayima, M . . . . . P16.02, P16.05 Naluyima, P . . . . . . . . . OA05.03 Namwat, C . . . . . . . . OA01.06 LB Nandi, A . . . . . . . . . . P19.33 LB Nanvubya, A . . . . . . . . . . P15.05 Naranbhai, V . . . . . . . . OA01.04 Nariya, S . . . . . . P18.28 LB, S07.01 Narpala, S . . . . . . . . . . . P05.06 Nassab, P . . . . . . . . . . . P08.04 Navis, M . . . . . OA02.04, OA06.05, P04.05 Nawaz, F . . . . . . . . . . . . P21.08 Nazarenko, O . . . . . . . . . P14.09 Ndhlovu, ZM . . . . . . . . OA06.01 Ndiaye, IP . . . . . . . . . P17.30 LB Ndungu, T . . . . OA01.04, OA05.02, P17.01, P20.05, P21.04 Ndure, J . . . . . . .OA09.06, P12.02 Negroni, M . . . . . . . . . . P20.01 Neidermyer, WJ . . . . . . . OA09.02 Newman, M . . . . . . . . . . P18.02 Newman, PA . . . . . P16.07, P16.09 Ngandu, N . . . . . OA06.04, OA07.02 Ngauy, V . . . . OA01.02, OA01.06 LB, OA07.06, OA07.08 LB, P14.13 LB Ngo-Giang-Huong, N . . . . . P21.03 Ngole, EM . . . . . . . . . . . P04.28 Nguay, V . . . . . . . . . . . . P01.03 Nichols, D . . . . . . . . . . . P07.02 Nielsen, L . . . . . . . P15.05, P16.06 Nielsen, M . . . . . . . . . . . P17.09 Nieuwenhuis, I . . . . . . . . P18.05

OConnor, DH . . . . . . . . OA08.04, P17.31 LB ODell, S . . . . . . OA04.04, P04.27, P04.42 LB, PL03.03 OGarra, A . . . . . . . P01.06, S05.02 OKeefe, B . . . . . . . . . . . P15.04 OMalley, J . . . . . . . . . . . P05.07 ONeal, T . . . . . OA04.02, P04.41 LB ORourke, SM . . . . .P04.33, P04.34 OConnell, R . . . . . . . OA01.06 LB, P14.13 LB ODell, S . . . . . . . . . . . OA07.03 OSullivan, A . . . . . . . . . . S07.01 Oballah, PO . . . . . . . . . . P04.13 Ochanda, J . . . . . . . . . . P09.09 Ochsenbauer, C . OA06.02, OA07.06, P09.15, P09.16 Ofek, G . . . . . . . . . . . OA04.03 Oh, S . . . . . . . OA03.01, P10.10 LB Oh, S . . . . . . . . . . . . . . P18.17 Oka, S . . . . . . . . . . . . . P17.08 Okocha, E . . . . . . . . . . OA08.03 Olonitola, SO . . . . . P20.05, P21.04 Olsen, OA . . . . . . . . . . . S02.05 Oniangue-Nzda, C . OA03.02, P08.03 Onlamoon, N . . . . . . . . . P17.27 Onyango, IA . . . . . . . . . . P09.09 Ortiz, A . . . . . . . . . . . OA08.01 Osanya, J . . . . . . . . . . . P12.02 Osawa, K . . . . . . . .P05.01, P19.03 Osman, N . . . . . . . P06.09, P06.10 Osmanov, S . . . . . . . . . . P16.04 Ostrowski, MA . . . . . . . P18.24 LB Ouma, BJ . . . . . . . . . . . P04.13 Ouzrout, B . . . . . . . . . . . P19.11 Overbaugh, J . . . . . . . . . OA07.07 Overman, G . . . . . . . . . . P12.01 Owens, J . . . . . . . . . . . OA04.03 Oyediran, K . . . . . . . . . . P15.10 Oyugi, J . . . . . . . . . . . . P09.09

236

AIDS Vaccine 2011

Author Index
Perez-Alvarez, S . . . . . . . . P06.06 Permar, S . . . . . . . . . . . P12.01 Perouzel, E . . . . . . . . . . . P02.05 Perreau, M . . . . . .OA01.01, P17.11, P19.20 Peters, B . . . . . . . . . . P18.21 LB Peters, H . . . . . . . . . . . . P18.12 Peterson, B . . . . . . . . . OA08.06 Peterson, ER . . . . . . . . . OA02.01 Phanuphak, N . . . . . . . P10.07 LB Phanuphak, P . . . . . . . P10.07 LB Phillips, E . . . . . . . . . . . P07.01 Phogat, S . . . . . OA03.03, OA04.05, P04.36 LB, P05.06, S02.05 Phuang-Ngren, Y . . . . . P10.07 LB Phung, B . . . . . . . . . . . . P14.01 Phung, P . . . . . . OA07.05, P04.33, P04.34, S02.05 Phuphuakrat, A . . . . . . . . P20.02 Pialoux, G . . . . . . . . . . . P14.01 Piatak, M . . . . . . . . . . . P19.27 Piatak, Jr, M . . . . . . . .OA11.06 LB Pichulik, T . . . . . . . . . . . P09.14 Picker, L . . . . . . . . . . . . P10.05 Pickeral, J . . . . . . . . . . . P09.16 Picking, RA . . . . . . . . . OA06.02 Picton, AC . . . . . . . . . . . P09.03 Piechocka-Trocha, A . . . . OA06.01, P07.05 LB, P09.13 Pilcher, C . . . . . . . . . . OA07.05 Pillay, M . . . . . . . . . . . . P17.01 Pillis, D . . . . . . . . . . . P20.08 LB Pilloto, JH . . . . . . . . . . . P04.08 Pilotto, J . . . . . . . . . . . . P20.03 Pinter, A . . . . . . . . . . . . P04.30 Pinyakorn, S . . . . P01.03, P10.07 LB Piovanetti, P . . . . . . . . . . P14.11 Pitisuttithum, P . . . . . OA07.08 LB, OA09.02, OA09.04, P14.03, PL01.02 Pitoiset, F . . . . . . . . . . . P19.05 Plana, M . . . P09.10, P11.04, P11.05, P14.10 Plotkin, S. . . . . . . . . . . SS01.01 Plummer, F . . . . P07.03 LB, P09.09, P17.03, P17.04, P17.05, P18.10, P18.12 Podzamczer, D . . . . . . . P18.21 LB Poignard, P . . . . OA04.01, OA04.05, OA10.05, P04.09, P04.42 LB, S02.05 Poizot-Martin, I . . . . . . . . P14.01 Pollara, J . . . . . . . P09.15, P09.16, P12.01 Pollard, R . . . . . . . . . . P18.21 LB Polonis, VR . . . . . OA07.06, P02.06, P04.13, P20.08 LB, P21.06 Polsrira, K . . . . . . . . . . . P17.27 Polyakov, N . . . P03.02 LB, P19.31 LB Poole, D . . . . . . . . . . . OA01.04 Poole, G . . . . . . . .P06.01, P06.02 Poon, A . . . . . . . . . . . . P08.04 Porichis, F . . . . . . OA06.01, P17.01 Potin, L . . . . . . . . . . . . P19.20 Power, K . . . . . . .OA03.02, P08.03 Prabakaran, P . . . . . . . . OA04.03 Premsri, N . . . . OA01.06 LB, P14.03, P14.13 LB Price, MA . . . . . P04.09, P20.09 LB

Qi, X . . . . . . . . . . . . P18.22 LB Qi, Y . . . . . . . . . . . . . . P08.03 Qi, Z . . . . . . . . . . . . P10.09 LB Qin, H . . . . . . . . . P01.06, S05.02 Qin, HR . . . . . . . . . . . OA04.03 Qin, Y . . . . . . . . . . . . OA10.01 Qiu, C . . . . . . . . . P17.17, P17.18 Qiu, S . . . . . . . . . P17.17, P17.18 Quakkelaar, ED . . . . P18.01, P18.05 Qureshi, H . . . . . . . . . P14.14 LB

R. Andre, RR . . . . . . . . P18.26 LB Radebe, MA . . . . . . . . . OA05.02 Rademeyer, C . . . . . . . P18.28 LB Raengsakulrach, B . . P06.12, P20.04 Rahman, M . . . . . . . . . . P08.04 Raimbault, M . . . . . . . . . P14.07 Rajakumar, P . . . . OA11.02, P03.01 Ramanathan, AA . . . . . . . P19.01 Ramesh, P . . . . . . . . . P19.33 LB Ramilo, O . . . . . . . P01.06, S05.02 Ramirez, N . . . . . . . . . . . P04.20 Ramos, A . . . . . . . . . . . S02.05 Ramsland, PA . . . . . . . . . P11.02 Ranasinghe, S . . OA05.01, OA07.05, P17.12 Ranchobe, N . . . . . .P04.29, P08.02 Rao, DS . . . . . . . . . . . OA04.06 Rao, M . . . . . OA02.05, OA07.08 LB, OA09.04, P02.06, P21.06 Rao, SS . . . . . . . . . . OA08.08 LB Rao, V . . . . . . . . . . . . . P02.06 Ratcliffe, SJ . . . . . . . . . OA08.01 Ratner, D . . . . . . . . . . P15.12 LB Rattanamanee, S . . . . . . . P01.03 Ratto-Kim, S . . . . . . . . OA01.02, P14.05, P18.06 Reardon, J . . . . . . . . . . . P09.13 Reddy, S . . .OA01.04, P11.01, P11.02 Reece, J . . . . . . . . . . . OA08.07 Reed, S . . . . . . . . P05.03, PL03.03 Reeves, K . . . . . . . . . OA11.06 LB Reeves, R . . . . . . OA11.02, P03.01, P19.27 Reilly, CS . . . . OA11.01, OA11.06 LB Reilly, M . . . . . . .OA09.06, P12.02 Ren, L . . . . . . . . . . . . . P04.22 Rerknimitr, R . . . P01.03, P10.07 LB Rerks-Ngarm, S . . . . . OA01.06 LB, OA06.03, OA07.08 LB, OA09.02, OA09.05, OA09.04, OA10.06, P14.03, P14.13 LB Reuter, M . . . . . . . . . . . P19.24 Rex, CG . . . . . . . . . . . P18.26 LB Reyes-Teran, G . . . . . . . P07.04 LB Riadi, G . . . . . . . . . . . . P07.01

Sacha, JB . . . . OA08.02, P18.24 LB Sagar, M . . . . . . . . . . . . P04.20 Sagi, Y . . . . . . . . . . . . . P04.15 Sahay, S . . . . . . . . . . P14.12 LB Sakarellos-Daitsiotis, M . . . OA03.04

AIDS Vaccine 2011

237

AUTHOR INDEX

Priddy, F . . . . . . P06.14 LB, P15.07 Prins, JM . . . . . . . . . . . . P04.06 Proundfoot, J . . . . . . . . OA06.01 Pruimboom-Brees, IM . . . P18.24 LB Pryluka, D . . . . . . . P17.02, P17.07 Pulendran, B . . . . . . . . . . S05.01 Pujol, F . . . . . . . . . . . . P06.06 Pumratana, K . . . . . . . . . P01.07 Pungpak, S . . . . . . . . . . P14.03 Purcell, D . . . . . . . P11.01, P11.02

Ribeiro, SP . . . . . . .P18.13, P18.18 Richert, L . . . . . . . . . . . P14.07 Richie, T . . . . . . . . . . . . P02.04 Richmond, M . . . . . . . . . P17.04 Ridtitid, W . . . . . P01.03, P10.07 LB Rieux, V . . . . . . . . . . . . P14.01 Rinaldo, CR . . . . . . . . . . P19.06 Riou, C . . . . . . . . . . . . . P17.20 Robb, M . . . . OA01.02, OA01.06 LB, OA05.03, P01.05, P01.07, P04.13, P06.05, P14.03, P14.05, P14.06, P14.13 LB, P20.07 LB, Robert-Guroff, M . . . . . . OA11.03, P07.02, P14.14 LB Roberts, JN . . . . . . . . . . OA11.03 Robertson, M . . . P14.14 LB, S07.01 Robinson, H . . . OA01.05, OA02.01, OA10.04, OA10.06, P02.08 Robinson, J . . . . . . P04.11, P04.30, P04.33, P04.41 LB, P08.01, P09.15 Rochas, M . . . . . . .P06.07, P18.20 Rochereau, N . . . . . . . . . P02.05 Rockstroh, JK . . . . . . . . P18.21 LB Rodrguez, A . . . . . P10.01, P17.02, P17.07, P18.07, P18.08 Roederer, M . . . . . . . . . OA07.03 Roesen, N . . . . . . . . . . . P18.17 Roger, T . . . . . . . . P19.18, P19.20 Rohan, L . . . . . . . . . . P15.12 LB Rolland, M . . . . . . . . OA08.08 LB, P01.02, P01.08, P18.28 LB, P19.22, S07.01, S07.02 Romain, G . . . . . . . . . . . P17.23 Rono, E . . . . . . . . . . . OA05.03 Rono, K . . . . . . . . . . . . P01.05 Rosa, DS . . . . . . . .P18.13, P18.18 Rosario, M . . . . . . . . . . . P18.01 Rosati, M . . . . . . . . . . OA10.04 Rose, N . . . . . . . . . . . . P18.15 Rosenberg, E . . . OA02.02, OA05.01, OA05.06, P09.07 Ross, A . . . . . . . . . . . . . P14.01 Rossenkhan, R- . . . . . . . . P12.03 Roungprakhon, S . . .P16.07, P16.09 Rountree, W . . . . . . . . . . P06.03 Routy, J . . . . . . . . . . . OA05.06 Roux, P . . . . . . . . . . . . P04.28 Rozehnal, J . . . . . . P06.07, P18.19, P18.20 Ruan, Y . . . . . . . OA06.06, P04.22 Rudersdorf, R . . . . . . . . OA08.02 Ruiz, MJ . . . . . . . . P17.02, P17.07 Ruiz-Riol, M . . . . . . . . . . P17.24 Rutvisuttinunt, W . . . . . . . P20.04 Ruzagira, E . . . . . . . . . . P15.07, P20.09 LB Rybicki, EP . . . . . . . . . . . P11.07 Rychert, J . . . . . OA05.01, OA05.06 Ryzhova, E . . . . . . . . . . OA08.01

Salazar-Gonzalez, J . . . . . . P08.01 Salemi, M . . . . . . . . . . OA05.04 Salisch, NC . . . . . . . . . . P03.01 Salmon-Cron, D . . . . . . . P14.07 Salomon, H . . . . . . P17.02, P17.07 Salomn, H . . . . . . . . . . P15.01 Sam, NE . . . . . . . . . . P04.39 LB Sambor, A . . . . . . . . . . . P06.03 Samleerat, T . . . . . . . . . . P21.03 Sampson, JM . . . . . . . . P04.41 LB Sanchez, A . . . . . . . . . . . P06.03 Snchez, J . . . . . . .P06.06, P19.08 Snchez, V . . . . . . . . . . . P17.24 Snchez-Merino, V . . . . . . P11.06 Snchez-Palomino, S . . . . . P11.04, P11.05 Sanders, EJ . . . . P04.09, P20.09 LB Sanders, RW . . . . . . . . . . P04.16 Sanders II, RC . . . . . . . . . P06.08 Sanders-Buell, E . . P20.07 LB, P20.08 LB, S07.01 Sandstrm, E . . . . OA09.05, P14.04, P14.06 Sanga, E . . . . . . . . . . . . P01.05 Sangare, K . . P06.07, P18.19, P18.20 Sangiamkittikul, A . . . . . . P20.04 Sant, A . . . . . . . . . . . . . P18.02 Sanz, I . . . . . . . . . . . . . P04.19 Saokhieo, P . . . . . . . . . . P06.04 Sardesai, N . . . . OA01.02, OA08.04, OA10.04, P05.08, P18.16, P18.25 LB Sardesai, N . . . . . . . . . . P19.01 Sarzotti-Kelsoe, M . . . . . . P06.03 Sastry, M . . . . . . . . . . OA03.03 Sato, A . . . . . . . . . . . . OA02.01 Sattentau, Q . . . . . . P02.09, P02.10 Savoye, A . . . . . . . . . . . . P17.11 Sayeed, E . . . . . . OA09.01, P18.06 Scadden, D . . . . . . . . . . P17.24 Schief, W . . . . . . . . . . OA03.03 Schiller, JT . . . . . . . . . . OA11.03 Schlessinger, S . . . . . . . . . S05.04 Schmidt, RE . . . . . . . . . . P09.05 Schmidt, S . . . .OA03.03, P04.36 LB, OA07.03, OA07.04, P04.23, P04.26, P04.27, PL03.01 Schneider, D . . . . . . . . . OA08.06 Schneidewind, A . . OA03.02, P08.03 Schramm, DB . . . . . . . . . P09.11 Schuetz, A . . . . . P01.03, P10.07 LB Schuitemaker, H . . . P04.06, P04.07, P04.16, P04.24 Schuman, J . . . . . . . . . . P05.07 Schutte, R . . . . . . . . . . OA04.03 Schwartz, O . . . . . . . . . . P09.12 Schweighardt, B . . . . . . . . P04.33 Scott, JK . . . . . . . . . . . OA04.03 Scott-Algara, D . . . . . . . OA02.03 Seaman, M . . . . OA04.02, OA07.01, OA10.02, P04.27, P05.03, P10.08 LB Sebunya, TK . . . . . . . . . . P12.03 Seder, RA . . . . . . . . . . . P17.22 Seeley, J . . . . . . . . . . . . P15.05 Seese, A . . . . . . . . . . . OA03.02 Seevers, S . . . . . . . . . . . P17.06 Sekaly, R . . . . . . .OA05.06, S05.04 Sekiziyvu, A . . . . . . . . . . P01.05 Self, S . . . . . . .P18.17, P18.23 LB, P18.27 LB

Author Index
Semeniuk, C . . . . . .P17.03, P18.10 SenGupta, D . . . . . . . . P18.24 LB Seoighe, C . . . . . . . . . . OA07.02, OA08.08 LB Sereti, I . . . . . . . . . . . . P01.03 Sette, A . . . . . . . . . . . . P17.12 Seung, E . . . . . . . . . . . . P17.15 Sexton, A . . . . . . . . . . OA08.07 Seydi, M . . . . . . . .P09.06, P21.05 Seymour, L . . . . . . . . . . . P18.17 Shahin, L . . . . . . . . . . . P12.03 Shahzad-ul-Hussan, S . . . OA03.03 Shalekoff, S . . . . . . . . . . P09.03 Shams-Rony, M . . . . . . . OA09.06 Shang, H . . . . . . . . . . . . P04.02 Shang, L . . . . . . . . . .OA11.06 LB Shanmugam, M . . . . . . . . P16.07 Shao, Y . . . . . . OA06.06, OA08.05, P04.22, P10.09 LB, P18.09 Shapiro, L . . . P04.37 LB, P04.40 LB Shapiro, R . . . . . . . . . . . P12.03 Sharma, M . . . . . . . P01.06, S05.02 Shattock, R . . . . . . P02.02, P10.03, P10.04, P10.06, P19.08, P19.14, P19.19, P21.01, S02.03 Shaw, BL . . . . . . . . . . P18.26 LB Shaw, GM . . . . . . P08.01, P09.15, S07.03 Shedlock, DJ . . . . . . . . . OA08.04 Shen, X . . . . . . . OA04.03, P19.01 Shephard, E . . . . . . P11.07, P18.11 Sheppard, NC . . . . . . . P18.24 LB Sherman, GG . . . . . . . . . . P09.11 Sherwat, A . . . . . P15.11, P18.25 LB Shete, A . . . . . . . . . . P14.12 LB Sheward, D . . . . . . . . OA04.07 LB, OA07.02, P04.03 Shi, X . . . . . . . P04.02, P04.38 LB Shianna, K . . . . . . . . . . . S03.03 Shiboski, S . . . . . . . . . P18.26 LB Shida, H . . . . . . . . . . . . P19.13 Shikuku, K . . . . . . . . . . OA05.03 Shin, T . . . . . . . . . . . . . P18.16 Shittu, OS . . . . . . .P20.05, P21.04 Shiver, J . . . . . . . . . . P14.14 LB Shmelkov, E . . . . . . . . . . P05.05 Shukair, S . . . . . . . . . . . S06.01 Sibeko, S . . . . OA01.04, OA04.07 LB, P04.03, P04.24 Siby Diallo, F . . . . . . . . . P06.07 Siddiqui, AA . . . . . . . . . . P10.03 Siebentritt, C . . . . . . . . . . P11.02 Sigirenda, S . . . . . . . . . . P16.06 Silbermann, B . . . . . . . . . P14.01 Silva, SY . . . . . . . . . . . . P05.04 Silvestri, G . . . . . . . . . . OA08.01 Simek, M . . . . . P04.09, P04.42 LB, P04.40 LB, S02.05 Simmons, A . . . . . . . . . . P09.14 Simmons, RP . . . . . . . . . P09.07 Sinangil, F . . . . . . P04.33, P04.34, P19.30 LB Singer, D . . . . . . . . . . P20.07 LB Sirivongrangson, P . . . . . . P06.12 Sirokman, K . . . . . . . . P19.33 LB Sitoe, N . . . . . . . . . . . . P06.09 Sivro, A . . . . . . . . . . . P07.03 LB Skinner, JA . . . . . . P01.06, S05.02 Skinner, PJ . . . . . . . . .OA11.06 LB Slack, C . . . . . . . . P16.07, S01.03 Slama, L . . . . . . . . . . . . P14.01 Sleasman, JW . . . . . . . . OA05.04 Slike, B . . . . . . . . . . . . . P01.05 Sloan, L . . . . . . . . . . . OA03.01 Smaoune, A . . . . . . . . . . P19.11 Smeaton, L . . . . . . . . . . P12.03 Smith, A . . . . OA11.01, OA11.06 LB, P15.12 LB Smith, KN . . . . . . . . . . . P19.06 Smith, LM . . . . . . . . . . . P19.26 Smith, R . . . . . . . . . . . OA06.03 Sobieszczyk, M . . . . P15.11, S04.03 Socias, ME . . . . . . . P17.02, P17.07 Soderberg, K . . . . OA06.02, P01.06, P09.16, S05.02 Soghoian, D . . . OA05.01, OA07.05, P17.12 Sokolovskiy, E . . . . . . . . . P14.09 Someya, K . . . . . . . . . . . P18.04 Sommerfelt, MA . . . . . . P18.21 LB Song, R . . . . . . . . . . . . OA07.01 Song, Y . . . . . . . . . . . OA03.06 Songsupa, R . . . . . . . . . . P06.04 Srensen, B . . . . . . . . P18.21 LB Soriano, V . . . . . . . . . . OA03.04 Sorzano, CS . . . . . .P14.10, P19.18 Soto-Nava, M . . . . . . . P07.04 LB Southern, Pj . . . . . . . .OA11.06 LB Sow, PS . . . . . . . . . . . . P21.05 Spearman, P . . . . . . . . . OA10.06 Spengler, M . . . . . . . . . OA10.03 Spentzou, A . . . . . . . . . OA09.01 Spies, G . . . . . . . . . . . . P10.05 Spire, B . . . . . . . . . . . . P14.01 Spotts, GE . . . . . . . . . . . P17.21 Spurrier, B . . . . . . . . . P04.41 LB Sriplienchan, S . . . . P01.07, P06.05 Srivastava, I . . . OA03.05 LB, P05.07 SS, P . . . . . . . . . . . . P20.06 LB Ssetaala, A . . . . . . . . . . . P15.05 Stablein, D . . . . . . . . . . . P14.03 Stamatatos, L . . . . . . . . . P05.07 Stambas, J . . . . . . . . . . OA08.07 Stanfield, RS . . . . . . . . OA04.05 Stebbings, R . . . . P17.28 LB, P18.15 Steel, G . . . . . . . . . . . . S04.04 Steinman, R . . . . . . . . . . S05.04 Stenescu, I . . . . . . . . . . . P19.08 Stephenson, KE . . P17.14, P19.30 LB Stephenson, S . . . . . . . P19.33 LB Sterjovski, J . . . . . . . . . . P11.02 Sternthal, S . . . . . . . . . . P15.09 Sterrett, S . . . . . . . . . . . S07.03 Stewart-Jones, G . . .P04.25, P19.07 Stieh, DJ . . . . . . . . . . . . P21.01 Stoddard, j . . . . . P18.28 LB, S07.01 Stone, GW . . . . . . . . . . . P19.22 Stratov, I . . . . . OA02.04, OA06.05, P04.05 Streaker, E . . . . . .OA04.03, P04.01 Streeck, H . . . . OA03.02, OA05.01, OA05.06, OA07.05, P17.12 Strokappe, N . . . . . . . . . P04.14 Stutz, H . . . . . . . . P11.07, P18.11 Su, B . . . . . . . . . . . . . OA07.04 Su, R . . . . . . . . . . . . P07.03 LB Subramanyam, T . . . . . . . P04.15 Sued, O . . . . . . . . P17.02, P17.07 Sukapirom, K . . . . . . . . . P17.27 Sukwit, S . . . . . . . . . . OA05.03 Sun, C . . . . . . . . . . . . . P19.17 Sun, Y . . . . . . OA05.04, P19.33 LB Sundling, C . . . . . . . . . OA04.04 Suntharasamai, P . . . . . . . P14.03 Surenaud, M . . . . . . . . . . P14.07 Susan, A . . . . . . . . . . P06.15 LB Sutthent, R . . . . . . . . . . P04.34 Suttichom, D . . . P01.03, P10.07 LB Suzuki, K . . . . . . . . . . . . P11.01 Swain, JV . . . . . . . . . . . P19.22 Swales, J . . . . . . . . . . . . P19.19 Swan, E . . . . . . . . . . . P10.08 LB Swetnam, J . . . . . . . . . . P05.05 Sylvester, A . . . . . . . . . . P19.24 Sylwester, A . . . . . . . . . . P10.05 Tomaras, G . . . . OA04.03, OA06.02, OA09.04, OA10.06, P04.24, P04.39 LB, P09.15, P09.16, P12.01, P17.16, S06.04 Tomescu, C . . . . . . . . . . . P14.11 Tong, T . . . . . . . . .P05.01, P19.03 Tongo, M . . . . . . . . . . . P04.28 Tongtoyai, J . . . . . . . . . . P20.04 Tornesello, M . . . . . P11.03, P19.15 Torrieri-Dramard, L . . . . . . P19.05 Toth, I . . . . . . . . . . . . . P09.13 Totrov, M . . . . OA10.02, P04.41 LB Tounkara, K . . . . . P06.07, P18.19, P18.20 Tovanabutra, S . . . . . . P20.07 LB, P20.08 LB, S07.01 Towle, T . . . . . . . . . . . P20.08 LB Tran, L . . . . . . . . . . . . . P04.24 Traore, S . . . . . . . . . . . . P06.07 Tregoning, JS . . . . . P10.04, P19.14 Treurnicht, F . . . . P17.20, P18.28 LB Trichavaroj, R . . . . . . OA01.06 LB, P01.03, P10.07 LB, P14.13 LB Trivett, MT . . . . . . . . . . OA08.06 Trubey, CM . . . . . . . . . OA08.06 Tsao, C . . . . . . . . . . . P04.39 LB Tsuji, M . . . . . . . . . . . . P02.04 Tucker, JL . . . . . . . . . . P06.14 LB Tucker, R . . . . . . . . . . P10.08 LB Tudor, D . . . . . . . OA10.03, S02.01 Tuff, J . . . . . . . . . . . . . P18.12 Tumba, N . . . . OA04.07 LB, P04.07 Tungsakul, V . . . . . . . . . . P06.05 Turk, G . . . . P17.02, P17.07, P18.08

AUTHOR INDEX

Tager, AM . . . . . . . . . . . P17.15 Tagliamonte, M . . . . P11.03, P19.15 Takamoto, K . . . . . . . . . OA10.01 Takawira, C . . . . . . . . . . P21.01 Takiguchi, M . . . . . . . . . . P17.08 Tamayo-Agrait, VM . . . . . . P14.11 Tan, J . . . . . . . . . . . . . . P11.02 Tan, Z . . . . . . . . . P15.06, P19.12 Tandon, R . . . . . . . . . P18.24 LB Tang, D . . . . . . . . . . . . P18.10 Tangy, F . . . . . . . .P09.12, P18.15 Tarosso, L . . . . . P17.25, P18.26 LB Tarragona, T . . . . . . . . . OA09.01 Tartaglia, J . . . . .OA01.01, OA01.03, OA01.06 LB, P14.13 LB, P19.25, SS01.02 Tarwater, P . . . . . . . . . P15.12 LB Tassaneetrithep, B . . . . . . . P01.05 Tataje, A . . . . . . . . . . . . P06.06 Tatoud, R . . . . . . . . . . . P19.19 Tatsuno, G . . . . . . . . . . . P04.34 Taylor, A . . . . . . . . . . . OA05.03 Tecleab, T . . . . . . . . . . OA09.05 Tekeste, S . . . . . . . . . . . P09.04 Tembe, N . . . . . . . P06.10, P06.09 Tepjan, S . . . . . . . .P16.07, P16.09 Termini, J . . . . . . . . . . . P19.22 Tewabe, N . . . . . . . . . . . P19.18 Thaitawat, N . . . . . P01.07, P06.05 Thakar, M . . . . . . . . . . P14.12 LB The IAVI African HIV Research Network, . . . . . . . . . . . P04.09 Thebus, R . . . . . . . . . . P18.28 LB Thenin, S . . . . . . . . . . . . P21.03 Thiebaut, R . . . . . . P01.06, P14.07, S05.02 Thienkrua, W . . . . . . . . . P06.12 Thior, I . . . . . . . . . . . . . P12.03 Thobakgale, CF . . . . . . . . P09.08 Thomas, EP . . . . . . . . . OA06.03 Thomas, M . . . . . . . . . P14.14 LB Thomas, V . . . . . . . . . . OA09.06 Thompson, MDiv, MS, SJ . . . P06.08 Thomson, H . . . . . . . . P06.16 LB Thongcharoen, P . . . . OA01.06 LB, P14.13 LB Thumbi, N . . . . . . . . . . . P12.03 Tian, M . . . . . . . . . . . . P18.09 Tian, X . . . . . . . . . . . . . P17.18 Tiberio, P . . . . . . . . . . . P19.04 Tichacek, A . . . P06.15 LB, P06.16 LB Tiemessen, CT . . . . . P09.03, P09.11 Tiraby, G . . . . . . . . . . . . P02.05 Tisserand, P . . . . . . . . . . P14.07 To, B . . . . . . . . . . . . . . P04.34

V, K . . . . . . . . . . . . . P14.12 LB Vabret, N . . . . . . . . . . . P09.12 Vaccari, M . . . . . OA01.03, P07.02, P19.25 Vaccine Immunology, N . . . P01.06, S05.02 Valentin, A . . . . . . . . . . OA10.04 Valenzuela-Ponce, H . . . . P07.04 LB Vallado, I . . . . . . . . . . . P19.14 Van den Dobbelsteen, M . . OA10.03 Van den Kerkhof, TL . . . . . . P04.16 Van der Stok, M . . . . . . . OA05.02 Van Engelenburg, F . . . . . OA10.03, S02.01 Van Gils, MJ . . . . . .P04.07, P04.16 Van Griensven, F . . . P06.12, P20.04 Van Gulck, E . . . . . . . . . . P17.23 Van Lunzen, J . . . . . . . P18.21 LB Van Ostade, X . . . . . . . . . P09.06 Van Roey, GA . . . . . . . . . P10.04 Van Ryk, D . . . . . . . . . . . P21.08 Vanham, G . . . . P04.25, P04.35 LB, P17.23 Vanijanonta, S . . . . . . . . . P14.03 Varangrat, A . . . . . . . . . . P06.12 Varughese, N . . . . . . . . . P15.09 Vasan, S . . . . . . P02.04, P17.29 LB VD, R . . . . . . . . . . . . P14.12 LB Veazey, R . . . . . . . . . . . P10.03 Veloso, V . . . . . . . . . . . . P04.08 Veloso, VG . . . . . . . . . . . P20.03 Venzon, D . . . . OA01.03, OA10.04, P07.02, P19.25

238

AIDS Vaccine 2011

Author Index
Vereecken, K . . . . . . . . P04.35 LB Verevochkin, S . . . . . . . . P14.09 Verma, A . . . . . . . . . . P14.12 LB Verrier, B . . . . . . . P02.05, P09.12, P19.08, P19.16 Verrips, T . . . . . . . . . . . . P04.14 Verwer, NL . . . . . . . . . . . P04.16 Viegas, EE . . . . . . .P06.09, P06.10 Vieira, VA . . . . . . . . . . . P17.25 Vigano, S . . . . . . . . . . . . P17.11 Villablanca, EJ . . . . . . . . . S06.03 Vine, S . . . . . . . . . . . . . . . . . OA06.01 Vinner, L . . . . . . . . . . . . P13.01 Vinton, CL . . . . . . . . . . . P17.22 Visarutratana, S . . . . . . . . P06.04 Visciano, M . . . . . . P11.03, P19.15 Vishwanathan, A . . . . . . . P21.07 Vogt, A . . . . . . . . . . . . P19.08 Vogt, J . . . . . . . . . . . . . P10.05 Von Briesen, H . . . . . . . . OA03.04 Von Gegerfelt, A . . . . . . OA10.04 Vorsatz, C . . . . . . . . . . . P20.03 Voss, G . . . . . . . . . . . . . P18.15 Vrbanac, V . . . . . . . . . . . P17.15 Weiqi, P . . . . . . . . . . . . P21.02 Weiss, H . . . . . . . . . . . PL02.01 Weiss, RA . . . . . . . . . . . P04.14 Weissenbacher, M . . . . . . . P15.01 Welles, HC . . . . . . OA01.01, P19.20 Wendel-Hansen, V . . . . . P18.21 LB Wesberry, M . . . OA07.06, P20.08 LB Westfall, D . . . . . . . . . . . S07.01 Whitney, JB . . . . . . . OA08.08 LB Wibmer, CK . . . OA04.07 LB, P04.29, P08.02 Wibmer, K . . . . . . . . . . OA07.02 Wieczorek, L . . . . . OA07.06, P02.06 Wietgrefe, S . . . . . . . .OA11.06 LB Wietgrefe, SW . . . . . . . . OA11.01 Wilkinson, P . . . . . . . . . . S05.04 Wilks, A . . . . . . . . . . . . P12.01 Willems, B . . . . . . . . . P04.35 LB Williams, C . . . . OA04.02, OA09.05, P04.41 LB Williamson, A . . . . OA11.05, P11.07, P18.11 Williamson, C . . . . . . OA04.07 LB, OA06.04, OA07.02, P04.03, P08.02, P17.20, P18.28 LB, PL02.02 Wilson, IA . . . . . .OA04.05, S02.05 Wilson, NA . . . . . . . . . OA08.02 Wilson, R . . . . . OA04.04, OA07.03 Wilton, S . . . . . . . . . . P07.05 LB Wimonsate, W . . . . . . . . . P06.12 Winchester, R . . . . . . . . . P09.11 Wininger, M . . . . . . . . P19.33 LB Wolf, E . . . . . . . . . . . . . P06.11 Wolfson, J . . . . . . . . . . . P01.08 Wong, CH . . . . . . . . . . . S02.05 Wong, FE . . . . . . . . . . . P17.10 Wong, K . . . . . . . . . . . . S07.01 Wongthanee, A . . . . . . . . P06.04 Woodman, ZL . . . . . . . . . P04.03 Woods, M . . . . . . . . . . . P09.01 Wren, L . . . . . . . OA02.04, P04.05 Wright, JK . . . . . . . . . . OA05.02 Wright, K . . . . . . . . . . . P19.04 Wrin, T . . . . . . . OA07.05, P04.09, P04.33, P04.34, S02.05 Wu, H . . . . . . . . . . . . . P17.16 Wu, L . . . . . . . . . . . . . PL03.01 Wu, X . . . . . . . OA04.01, OA07.03, P04.27, P04.37 LB, P04.39 LB, P04.42 LB, P04.40 LB Wu, Z . . . . . . . . . . . . . P15.06 Wyatt, L . . . . . . . .P02.08, P19.09 Wyatt, R . . . . . OA04.04, OA07.03, OA10.05, P04.18, P05.06, P19.32 LB Xu, Z . . . . . . . . . . P01.06, S05.02 Xue, Y . . . . . . . . . . . . OA03.01 Yamamoto, N . . . . . . . . . P18.04 Yamkram, T . . . . . . . . . . P06.05 Yan, H . . . . . . . . . . . . . P17.16 Yan, J . . . . . . . OA01.02, OA08.04, P05.08, P18.16, P19.22, P19.24 Yan, J . . . . . . . . . . . . . P19.01 Yang, H . . . . . . . . . . . . P17.16 Yang, L . . . . . . .OA03.06, OA04.06 Yang, Y . . . . . . OA03.03, OA04.03, P04.36 LB, P04.42 LB Yang, Z . . . OA03.03, P04.24, P04.27 Yang, ZY . . . . . . . . . . . PL03.01 Yao, C . . . . . . . . . . . P04.38 LB Yarbrough, K . . . . . . . . OA02.03 Yassine-Diab, B . . . . . . . OA05.06 Yates, N . . . . . . . . . . . . P12.01 Yim, S . . . . . . . . . . . . . P16.09 Yin, J . . . . . . . . . . . . . OA08.04 Yin, L . . . . . . . . . . . . . OA05.04 Yinfeng, Z . . . . . . .P17.26, P21.02 York, VM . . . . . . . . . . P18.26 LB Yu, X . . . . . . . . . . . . . OA06.03 Yu, Y . . . . . . . . . . . . . . P17.10 Yuan, M . . . . . . . . . . . . P19.04 Yuan, S . . . . P17.17, P17.18, P19.24 Yuan, T . . . . . . . . .P04.01, P04.12 Yuan, X . . . . . . . . . . . . P18.10 Yue, L . . . . . . . . . . . . . P08.01 Yuste, E . . . . . . . . . . . . . P11.06

Wabwire-Mangen, F . . . . . P04.13 Wachihi, C . . . . . . .P17.03, P18.12 Wachihi, C . . . . . . . . . . . P17.04 Waelti Da Costa, V . . . . . . P20.01 Wagner, D . . . . . . . . . . . P05.06 Wagner, R . . . . . OA01.01, P04.10, P19.08 Wahren, B . . . . . OA09.05, P14.04, P14.06 Wakefield, SF . . . . . P06.08, S01.05 Walker, B . . . . . OA02.02, OA03.02, OA05.01, OA05.02, OA05.06, OA06.01, P07.05 LB, P08.03, P08.04, P09.02, P09.08, P17.01, P17.12, P17.24 Walker, LM . . . . OA04.05, OA10.05, P05.06, S02.05 Wallace, S . . . . . . . . . . . P15.08 Walsh, P . . . . . . . . . . . OA10.06 Walsh, S . . . . . . . . . . P10.08 LB Wan, D . . . . . . . . . . . . . P19.07 Wan, Y . . . . . . . . . . . . . P18.03 Wang, H . . . . . . P04.38 LB, P19.12 Wang, R . . . . . OA05.05, P10.09 LB Wang, S . . . . . . OA10.02, P04.31, P10.09 LB, S06.03 Wang, SK . . . . . . . . . . . S02.05 Wang, W . . . . . . . .P17.17, P17.18 Wang, X . . . . . . .OA04.02, P04.02 Wang, Y . . OA04.03, P04.01, P14.02 Wang, YE . . . . . . . . . . . P08.03 Warren, M . . . . . . .P16.04, P16.08 Waterman, D . . . . . . . OA11.06 LB Watkins, DI . . . . OA08.02, OA08.06 Wayengera, M . . . . . . . . . P19.02 Weber, J . . . . . . . . . . . . P19.19 Wegmann, F . . . . . . P02.09, P02.10 Wei, D . . . . . . . . . . . . . P21.08 Weijtens, M . . . . . . . . P10.08 LB Weiner, D . . . . . . OA08.04, P05.08, P18.16, P18.25 LB , P19.01, P19.22, P19.24 Weinhold, KJ . . . . . . . . OA06.02

Xiao, J . . . . . . . . . . . . . P17.17 Xiao, W . . . . . . . . . . . . P19.09 Xiao, X . . . . . . . . . . . . . P14.02 Xie, J . . . . . . . . . . . . P07.03 LB Xing, L . . . . . . . . . . OA03.05 LB Xu, J . . . . . . . . . .P17.17, P17.18 Xu, K . . . . . . . . . . . . . OA07.04 Xu, L . . . . . . . . . . . . . PL03.01 Xu, X . . . . . . . . . . . . . . P17.16 Xu, Y . . . . . . . . P17.32 LB, P19.12

Zak, DE . . . . . . . . . . . OA02.01 Zakareishvili, N . . . . . . . . P15.03 Zambonelli, C . . . . . . . P19.33 LB Zaunders, JJ . . . . . . . . P17.32 LB Zeng, M . . . . OA11.01, OA11.06 LB Zhan, W . . . . . . . . . . P18.24 LB Zhang, A . . . . . . . .P17.17, P17.18 Zhang, B . . . . P04.36 LB, P04.37 LB, P04.42 LB, P04.40 LB Zhang, C . . . . . . . . . . . . P14.02 Zhang, H . . . . . . . . . . . . P19.28 Zhang, L . . . . . .P04.02, P04.38 LB, P19.17 Zhang, M . . . . . . . P04.01, P04.12 Zhang, X . . . . . . . .P17.17, P17.18 Zhang, Y . . . . . . . . . . . . P04.01 Zhao, C . . . . . . . . . . . . P04.22 Zhao, H . . . . . . P18.28 LB, S07.01 Zhao, Q . . . . . . . OA04.03, P17.06 Zheng, B . . . . . . . . . . . . P04.19 Zheng, Y . . . . . . . . . . . . P04.22 Zhou, J . . . . . . . . . . . . . P19.12 Zhou, M . . . . . . .OA03.04, P17.17 Zhou, P . . . . . . . . . . . OA03.06 Zhou, T . . . . P04.37 LB, P04.40 LB, PL03.01 Zhu, J . . . . . P04.37 LB, P04.42 LB, P04.40 LB Zhu, P . . . . . . . . . . . . OA03.06 Zhu, Z . . . . . . . .OA04.03, P14.02 Zolla-Pazner, S . . OA04.02, OA09.05, OA09.04, OA10.02, P04.30, P04.32, P04.41 LB, P05.05

AIDS Vaccine 2011

239

AUTHOR INDEX

Zorrilla, CD . . . . . . . . . . . P14.11 Zuo, T . . . . . . . . . . . . P04.38 LB Zupancic, M . . . . . . . .OA11.06 LB Zupkosky, J . . . . . . . . . . P17.01 Zurawski, G . . . OA03.01, P10.10 LB Zurawski, S . . . OA03.01, P10.10 LB Zurbriggen, R . . . . . . . . OA10.03

Notes

240

AIDS Vaccine 2011

Notes

AIDS Vaccine 2011

241

Notes

242

AIDS Vaccine 2011

Notes

AIDS Vaccine 2011

243

Notes

244

AIDS Vaccine 2011

Notes

AIDS Vaccine 2011

245

Notes

246

AIDS Vaccine 2011

Tear out certificate along perforation.

Certificate of Attendance
This document is to certify that

AIDS Vaccine 2011 Conference

attended the AIDS Vaccine 2011 Conference held September 12-15 at the Bangkok Convention Centre, Bangkok, Thailand.

Pratap Singhasivanon

Conference Chair

Punnee Pitisuttithum
Conference Co-Chair

Manit Teeratantikanont
Conference Co-Chair

Bangkok Convention Centre at CentralWorld


http://www.vaccineenterprise.org/

Das könnte Ihnen auch gefallen