19th Annual International Meeting of the Institute of Human Virology
1
Contents
CONTENTS
Click on any content title to link to that section.
03
Program Information
and Acknowledgements
14
Events Schedule
17
Speaker Schedule
25
Abstract Index
30
Poster Index
19th Annual International Meeting of the Institute of Human Virology
2
Program Information and
Acknowledgements
Welcome
19th Annual International Meeting of the Institute of Human
Virology at the University of Maryland School of Medicine
Dear Colleagues and Friends,
You are invited to join us for the 19th Annual International Meeting of the Institute of Human Virology (IHV) scheduled for
Monday, October 23, 2017 through Thursday, October 26, 2017 at the Four Seasons Hotel in Baltimore, Maryland. This year,
the Annual Meeting will feature presentations on the latest advances in: HIV “Cure” Research, Preventative and Therapeutic
Vaccines, Immunology and Viral Pathogenesis, Viral Diagnostics, Emerging Concepts in Cancer Therapy, Cancer and
Stem Cells, Infectious Agents and Cancer, Public Health Science and Responses - From Local to Global, Clinical Virology Cardiovascular and Liver Complications of Viral Infections, Junior investigators are invited to submit research abstracts for
oral or poster presentation, so please share this opportunity with your faculty and colleagues.
A special mini-symposium will honor this year’s IHV Lifetime Achievement Awardees: Lifetime Achievement Award for
Public Service: Quarraisha Abdool Karim, PhD Associate Scientific Director, Centre for the AIDS Program of Research
in South Africa (CAPRISA), Salim S. Abdool Karim, MBChB, PhD, DSc Professor for Global Health, Department of
Epidemiology, CAPRISA; Lifetime Achievement Award for Scientific Contributions: Peter Palese, MD Horace W.
Goldsmith Professor of Medicine, Chair of the Department of Microbiology, Professor of Microbiology and Medicine, Icahn
School of Medicine at Mount Sinai
In addition to other prominent presenters, this symposium will feature the fourth annual Reinhard Kurth Memorial Lecture
by Peter Palese, MD. The Annual Awards Gala will be held Wednesday, October 25 at the Four Seasons Hotel, Baltimore.
A gala reception will begin at 6:00 p.m. followed by dinner at 7:00 p.m. We look forward to welcoming you to Baltimore this
October as we continue our annual tradition of excellent science and provocative discussion.
Sincerely,
Robert C. Gallo, MD
Manhattan Charurat, PhD
Homer and Martha Gudelsky
Director, Division of Epidemiology and Prevention
Distinguished Professor in Medicine
Professor of Medicine, Institute of Human
Director, Institute of Human Virology
Virology
Co-Founder and Director, Global Virus Network
19th Annual International Meeting of the Institute of Human Virology
3
Program Information and
Acknowledgements
MISSION STATEMENT
The Institute was established to create and develop a world-class center of excellence focusing on chronic viral diseases,
especially HIV/AIDS, and virally-linked cancers.
The IHV is dedicated to the discovery, research, treatment and prevention of these disease.
Its unique structure seeks to connect cohesive, multi-disciplinary research and clinical programs so that new treatments
are streamlined from discovery to patient. The IHV serves patients locally and the scientific community globally.
Robert C. Gallo, MD
Director
Robert R. Redfield, MD
Associate Director
Director, Division of Clinical Care and Research
Man E. Charurat, PhD
Director, Division of Epidemiology and Prevention
George Lewis, PhD
Director, Division of Vaccine Research
Wuyuan Lu, PhD
Co-Director, Division of Basic Science
Eric Sundberg, PhD
Co-Director, Division of Basic Science
Dave Wilkins
Chief Operating Officer
19th Annual International Meeting of the Institute of Human Virology
4
Program Information and
Acknowledgements
Institute of Human Virology (IHV) Board of Advisors
Terry Lierman (Chair)
Sheilah Kast
On the Record, WYPR
The Honorable Catherine E. Pugh
Mayor of Baltimore
Ellen Ratner
Talk Radio News Service
William A. Blattner, MD
Institute of Human Virology, Retired
John R. Kelly
Kelly & Associates Insurance Group
(KELLY)
KELLY Benefit Strategies & KELLY
Advisory
Robert Charrow, Esquire
Greenberg-Taurig, LLP
William E. “Brit” Kirwan, PhD
University System of Maryland
Barbara J. Culliton
Science Journalist and Policy
Consultant
The Honorable Nancy Kopp
Maryland State Treasurer
The Honorable Kathleen Kennedy
Townsend
The Rock Creek Group
Peter Angelos, Esquire
The Law Offices of Peter Angelos,
Esquire
The Honorable Elijah Cummings
United States House of
Representatives
John Evans
Evans Telecommunications
The Honorable Arthur J. Gajarsa
WilmerHale
Michael Greenebaum
Greenebaum Enterprises
Harry Huge
Harry Huge Law Firm LLP
Mark H. Kaplan, MD
University of Michigan Health System
John McHutchison, MD
Gilead Sciences, Inc.
Charles R. Modica, MD
Saint George’s University
Timothy Moynahan, Esquire
The Moynahan Law Firm, LLC
Guangqi Tian
Sino Invest Limited
Jeffrey B. Trammell
Trammell and Company
Lenny Wilkens
National Basketball Association
Hall of Fame Coach and Player
Danny Wong
Medisun
Franco Nuschese
Georgetown Entertainment Group
Ambassador R. James Woolsey
Venture Partner, Lux Capital
Management
Joseph S. Pagano, MD
University of North Carolina
Steve Wozencraft
John O’Donnell Associates, LLC
Peter Palese, PhD
Mount Sinai School of Medicine
HONORARY
James Pinkerton
RATE Coalition
19th Annual International Meeting of the Institute of Human Virology
Stewart Greenebaum
The Honorable Ernest F. Hollings
Ray Lewis
Princess Chulabhorn Mahidol
5
Program Information and
Acknowledgements
IHV Scientific Advisory Board
Joseph Pagano, MD (Chair)
University of North Carolina
Edward Berger, PhD
NIAID/NIH
Willi K. Born, PhD
National Jewish Health
William DeGrado, PhD
University of California, San Francisco
Max Essex, DVM, PhD
Harvard AIDS Institute
Warner C. Greene, MD, PhD
Gladstone Institute of Virology and
Immunology
Michael S. Saag, MD
University of Alabama at Birmingham
Center for AIDS Research
Mark H. Kaplan, MD
University of Michigan Health System
Erica Ollmann Saphire, PhD
The Scripps Research Institute
Michel Klein, MD
VaxiBio, Inc.
Mario Stevenson, PhD
University of Miami
Douglas Nixon, MD, PhD
The George Washington University
Sten Vermund, PhD
Vanderbilt University
Erling C. J. Norrby, MD, PhD
Secretary General, The Royal
Swedish Academy of Sciences
19th Annual International Meeting of the Institute of Human Virology
6
Program Information and
Acknowledgements
Organizing Committees
The Institute of Human Virology at the University of Maryland School of Medicine is grateful for the assistance
provided by our International and Local Organizing Committees.
International Organizing Committee
Robert C. Gallo, MD, Chair
Director, Institute of Human Virology
Guido Poli, MD,
San Raffaele Scientific Institute
Sharon Lewin, FRACP, PhD,
The University of Melbourne
Richard Wyatt, PhD,
The Scripps Research Institute
Local Organizing Committee
Robert C. Gallo, MD, Chair
Director, Institute of Human Virology
Wuyuan Lu, PhD,
Institute of Human Virology
Man Charurat, PhD, Co-Chair
Fabio Romerio, PhD,
Institute of Human Virology
Eric Sundberg, PhD,
Institute of Human Virology
Anthony DeVico, PhD,
Institute of Human Virology
Shyamasundaran Kottilil, MD, PhD,
Institute of Human Virology
19th Annual International Meeting of the Institute of Human Virology
Davide Zella, PhD,
Institute of Human Virology
Chozha V. Rathinam, PhD,
Institute of Human Virology
7
Program Information and
Acknowledgements
Communications and Press Policy
To enhance the exchange of information and communication among attendees of the Institute of Human Virology Annual
International Meeting, the following must be adhered to by all participants:
•
No coverage, reporting or publication of scientific data or presentations at the Institute
of Human Virology Annual Meeting is permitted without the written consent of the
presenter(s) and Nora Grannell (info below). This rule applies to all forms of media,
including blogging, tweeting, etc.
•
Alternatively, if the content does not contain information about non-published data,
or comments made during the closed meeting, all forms of media are acceptable
without written consent.
One-on-one interviews with scientists and media may be arranged by contacting Nora Samaranayake, Director of Public
Relations and Marketing, Institute of Human Virology, (410) 706-1954 or NSamaranayake@ihv.umaryland.edu.
Those registering for the meeting as “press” must provide their credentials within 3 days to
ihvmeeting@ihv.umaryland.edu.
19th Annual International Meeting of the Institute of Human Virology
8
Program Information and
Acknowledgements
Special Acknowledgements
The Institute of Human Virology at the University of Maryland would like to thank the following organizations. Without their
continued and generous support, this meeting would not be possible.
National Institute of Allergy and Infectious Diseases*
Division of AIDS, NIH
Gilead
National Institute on Drug Abuse
Sanofi Pasteur
Profectus BioSciences, Inc.
Merck
Abbott Molecular
The Marlene and Stewart Greenebaum Cancer Center
Ortho Clinical Diagnostics
*Funding for this conference was made possible, in part, by 5 R13 AI 046078 - 18 from the National Institute of
Allergy and Infectious Diseases. The views expressed in written conference materials or publications and by speakers
and moderators do not necessarily reflect the official policies of the Department of Health and Human Services; nor
does mention of trade names, commercial practices, or organizations imply endorsement by the U.S. Government.
19th Annual International Meeting of the Institute of Human Virology
9
Program Information and
Acknowledgements
The 2017 IHV Lifetime Achievement Award
for Scientific Contributions
Peter Palese, PhD
Dr. Peter Palese is a Professor of Microbiology and Medicine, and the Chair of the Department
of Microbiology at the Icahn School of Medicine at Mount Sinai. His research is in the area
of RNA-containing viruses with a special emphasis on influenza viruses. He established
the first genetic map for influenza A, B, and C viruses, identified the function of several
viral genes, and defined the mechanism of neuraminidase inhibitors (which are now FDAapproved antivirals). He was also a pioneer in the field of reverse genetics for negative
strand RNA viruses. His laboratory’s research is currently focused on the development of a
universal influenza virus vaccine and oncolytic viruses.
Palese is a member of the National Academy of Sciences, the National Academy of Medicine
(formerly IOM), and the American Academy of Arts and Sciences. He is also a corresponding
member of the Austrian Academy of Sciences and a member of the German Academy of
Sciences (Leopoldina). Palese is a recipient of the Robert Koch Prize, the Sanofi-Institut
Pasteur Award, the Beijerinck Virology Prize, and the Maurice Hilleman/Merck Award; he has
received honorary doctorate degrees from The Mount Sinai School of Medicine, Baylor College of Medicine and McMaster
University. Dr. Palese serves on the editorial board for the Proceedings of the National Academy of Sciences and is a former
president of the Harvey Society and the American Society for Virology.
19th Annual International Meeting of the Institute of Human Virology
10
Program Information and
Acknowledgements
The 2017 IHV Lifetime Achievement Award
for Public Service
Quarraisha Abdool Karim, PhD
Quarraisha Abdool Karim, PhD, Associate Scientific Director of CAPRISA, is an infectious
diseases epidemiologist whose main research interests are in understanding the evolving
HIV epidemic in South Africa; factors influencing acquisition of HIV infection in adolescent
girls; and sustainable strategies to introduce antiretroviral therapy in resource-constrained
settings. She holds Professorships in Clinical Epidemiology at the Mailman School of Public
Health, Columbia University, USA and in Public Health at the Nelson R Mandela School of
Medicine, University of KwaZulu-Natal in South Africa. She is also a visiting scientist at
Massachusetts General Hospital and Visiting Lecturer at Harvard University. Since 1998 she
has played a central role in building the science base in southern Africa through the Columbia
University - Southern African Fogarty AIDS International Training and Research Programme
that has trained over 600 scientists in southern Africa.
She was the Principal Investigator of the landmark CAPRISA 004 tenofovir gel trial which
provided proof of concept for Microbicides, highlighted by Science as one of the Top 10
scientific breakthroughs in 2010. Professor Abdool Karim’s has over 170 peer reviewed publications and has authored
several books and book chapters.
Professor Abdool Karim is currently chair of the South African National AIDS Council Prevention Technical Task Team,
a member of the UNAIDS Scientific Expert Panel and Scientific Advisor to the Executive Director of UNAIDS. She is an
advisory board member of the Higher Education and Training HIV/AIDS Programme (HEAIDS), Scientific Advisory Board
member of the US President’s Emergency Pan for AIDS Relief (PEPFAR), Chair of the PEPFAR Adolescent Girls and Young
Women Expert Working Group, a member of the HIV Centre Strategic Advisory Committee and the NIH OAR Microbicides
Planning Group. She is a Foreign Associate member of the US US National Academy of Medicine, Fellow of the Royal
Society of South Africa, Fellow of the Academy of Science of South Africa, Fellow of the African Academy of Sciences and
Fellow of The World Academy of Sciences (TWAS). She is currently Vice-President (Southern African Region) of the African
Academy of Sciences.
19th Annual International Meeting of the Institute of Human Virology
11
The 2017 IHV Lifetime Achievement Award
for Public Service
Salim S. Abdool Karim, MBChB, PhD, DSc
Salim S. Abdool Karim, MBChB, PhD, DSc (honoris causa) is a clinical infectious diseases
epidemiologist widely recognised for his ground-breaking scientific contributions in HIV
prevention and treatment. He is Director of CAPRISA - Centre for the AIDS Programme of
Research in South Africa; Pro Vice-Chancellor (Research), University of KwaZulu-Natal;
CAPRISA Professor of Global Health at Columbia University, Adjunct Professor of Medicine,
Cornell University and Associate Member of The Ragon Institute of Massachusetts General
Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard University.
His contributions to microbicides for HIV prevention spans two decades and culminated in
the CAPRISA 004 tenofovir gel trial which provided proof-of-concept that antiretroviral drugs
can prevent sexually transmitted HIV infection and herpes simplex virus type 2 in women.
He is co-inventor on patents which have been used in several HIV vaccine candidates and
his clinical research on TB-HIV treatment has shaped international guidelines on the clinical
management of co-infected patients.
He chairs the UNAIDS Scientific Expert Panel and is a member of both the WHO HIV-TB Task Force and the WHO Expert
Panel on STIs and HIV. He is an elected Fellow of the World Academy of Sciences, African Academy of Sciences, Academy
of Science in South Africa, Royal Society of South Africa and American Academy of Microbiology. He is a Foreign Associate
Member of the US National Academy of Medicine. He serves on the Boards of Lancet-Global Health, Lancet-HIV and the
New England Journal of Medicine.
19th Annual International Meeting of the Institute of Human Virology
12
Program Information and
Acknowledgements
Previous Recipients of IHV Lifetime Achievement Awards
LIFETIME ACHIEVEMENT AWARDS FOR SCIENTIFIC CONTRIBUTIONS
1999
George Klein, MD, Karolinska Institute, Stockholm, Sweden
2000
Maurice Hilleman, PhD, Merck Research Laboratories, Sumneytown, Pennsylvania, USA
2001
Hilary Koprowski, MD, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
2002
Alexander Rich, MD, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
2003
Jan Svoboda, PhD, DSc, Institute of Molecular Genetics, Prague, Czech Republic
2004
Paul Zamecnik, MD, Massachusetts General Hospital, Boston, Massachusetts, USA
2005
Manfred Eigen, PhD, Max Planck Institute, Göttingen, Germany
2006
Maxine Singer, PhD, National Institutes of Health, Bethesda, Maryland, USA
2008
Isaac P. Witz, PhD, Tel Aviv University, Tel Aviv, Israel
2010
Rino Rappuoli, PhD, Novartis Vaccines, Sienna, Italy
2011
Max Essex, DVM, PhD, Harvard AIDS Institute, Boston, Massachusetts, USA
2012
Thomas A. Waldmann, MD, National Cancer Institute, Bethesda, Maryland, USA
2013
Vadim I. Agol, MD, PhD, DSc, Russian Academy of Medical Sciences, Moscow, Russia
2014
William Paul, MD, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
2015
Harald zur Hausen, MD, Nobel Laureate, Gelsenkirchen, Germany
2016
Peter Vogt, Ph.D., Scripps Research Institute, La Jolla, California, USA
LIFETIME ACHIEVEMENT AWARD FOR PUBLIC SERVICE
2004
Stewart Greenebaum, Greenebaum and Rose Associates, Inc., Baltimore, Maryland, USA
2006
Martin Delaney, Project Inform, San Francisco, California, USA
2008
John D. Evans, Evans Telecommunication Company, Miami, Florida, USA
The Honorable Robert K. Gray, Gray and Company II, Miami, Florida, USA
2010
Harry Huge, Esq., The Harry and Reba Huge Foundation, Charleston, South Carolina, USA
2011
Bernadine Healy, MD, In Memoriam, Former Director National Institutes of Health, Bethesda, MD, USA
2012
Yi Zeng, PhD, China Centers for Disease Control, Beijing, China
2013
José G. Esparza, MD, PhD, Bill & Melinda Gates Foundation, Seattle, Washington, USA
2014
John Martin, PhD, Gilead Sciences, Inc., Foster City, California, USA
2015
Anthony S. Fauci, MD, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
2016
Ray Schinazi, Ph.D., Hon DSc, Emory University, Atlanta, Georgia, USA
ONE-TIME LIFETIME ACHIEVEMENT AWARD FOR EXCELLENCE IN TEACHING
2010
Michele LaPlaca, MD, Institute of Microbiology of the University of Bologna, Bologna, Italy
LIFETIME ACHIEVEMENT AWARD FOR EXCELLENCE IN MEDICAL EDUCATION, CLINICAL CARE AND CLINICAL RESEARCH
2012
John G. Bartlett, MD, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
19th Annual International Meeting of the Institute of Human Virology
13
Events Schedule
Evening Events Schedule
Monday, October 23, 2017
Opening Reception
Tuesday, October 24, 2017
Poster Session
6:30 – 8:30 pm
6:10 – 8:30 pm
Grand Prefunction
Grand Prefunction
Wednesday, October 25, 2017
6:00 – 7:00 pm
7:00 pm
19th Annual International Meeting of the Institute of Human Virology
Lifetime Achievement Awards
Gala Reception
Grand Prefunction
Lifetime Achievement Gala
Banquet and Awards Ceremony
Cobalt Ballroom
14
Events Schedule
Program Overview
Monday, October 23, 2017
8:30 am – 12:20 pm
Session A - HIV “Cure” Research
10:10 – 10:25 am
Coffee Break
12:20 – 1:35 pm
Lunch Break
1:35 – 3:40 pm
Session B – Emerging Concepts in
Cancer Therapy
3:40 – 3:55 pm
Coffee Break
3:55 – 6:25 pm
Session C – Preventative and
Therapeutic Vaccines
6:30 – 8:30 pm
Opening Reception
Tuesday, October 24, 2017
8:30 am – 12:05 pm
10:10 – 10:25 am
12:05 – 1:20 pm
1:20 – 2:35 pm
Session D – Immunology and Viral
Pathogenesis
Coffee Break
Lunch Break
Session D – Immunology and Viral
Pathogenesis (continued)
2:35 – 4:15 pm
Session E – Public Health Science and
Responses - From Local to Global
4:15 – 4:30 pm
Coffee Break
19th Annual International Meeting of the Institute of Human Virology
15
Events Schedule
Program Overview, continued
4:30 – 6:10 pm
Session F – Cancer and Stem Cells
6:10 – 8:30 pm
Poster Session
Wednesday, October 25, 2017
8:30 am – 12:05 pm
Session G – Infectious Agents and
Cancer
10:10 – 10:25 am
Coffee Break
12:05 – 2:15 pm
Lunch Break
2:15 – 5:55 pm
Session H – Lifetime Achievement
Award Mini-Symposium
4:00 – 4:15 pm
Coffee Break
6:00 – 7:00 pm
Gala Reception
7:00 pm
Lifetime Achievement Awards Banquet
Thursday, October 26, 2017
8:30 am - 12:30 pm
Session I – Advances in Clinical
Virology - Cardiovascular and Liver
Complications of Viral Infections
10:10 – 10:25 am
Coffee Break
12:05 – 1:05 pm
Lunch Break
1:05 – 3:30 pm
3:30 pm
19th Annual International Meeting of the Institute of Human Virology
Session J- Special Symposium on
Advances in Viral Diagnostics
Adjourn
16
Speaker Schedule
Speaker Schedule
Monday, October 23, 2017
Session A:
HIV “Cure” Research
Grand Ballroom
Chairpersons and Discussants:
Diana Finzi, PhD, National Institutes of Health
Robert Siliciano, MD, Johns Hopkins University
Carl Dieffenbach, PhD, DAIDS
8:30
Monsef Benkirane, PhD, University of Montpellier
Understanding for targeting the HIV reservoir
A-101
8:55
Guido Ferrari, MD, Duke University
Antibody binding to HIV-1 infected cells as mechanism for treatment of HIV infection
A-102
9:20
Mirko Paiardini, PhD, Emory University
A-103
Combined immune-based strategies targeting viral persistence in ART-suppressed SIV-infected
rhesus macaques
9:45
Douglas Nixon, MD, PhD, George Washington University
Immune Targeting of the Latent Reservoir
A-104
Coffee Break, 10:10 AM - 10:25 AM Grand Prefunction
10:25
Mary Kearney, PhD, National Cancer Institute
A-105
No evidence for HIV Replication in lymph nodes during ART
10:50
Kevin Morris, PhD, Beckman Research Institute of the City of Hope
A-106
Eradication of HIV; molecular therapeutic paths to a functional cure
11:15
Jintanat Ananworanich, MD, PhD, U.S. Military HIV Research Program
Early ART and HIV Remission: Experience from the RV254 and related HIV remission studies
A-107
11:40
Deanna Kulpa, PhD, Emory University
The Contribution of Memory CD4+ T cell subset phenotype to latency reversal efficiency
A-108
12:05
Early Stage Investigator: Ya-Chi Ho, MD PhD, Johns Hopkins University
A-109
Cytotoxic T lymphocytes shape the landscape of HIV-1 proviruses
Lunch Break, 12:30 PM – 1:35 PM
19th Annual International Meeting of the Institute of Human Virology
17
Speaker Schedule
Session B:
Emerging Concepts in Cancer Therapy
Grand Ballroom
Chairpersons and Discussants:
Eduardo Sotomayor, MD, George Washington University
Isaac Witz, PhD, Tel Aviv University
B-101
1:35
Yutaka Tagaya, PhD, Institute of Human Virology
Shooting Many Cytokines with a Single Stone - a Novel Anti-cytokine Technology with
Broad Clinical Application
1:55
Paul Frohna, MD, PhD, PharmD, Bioniz Therapeutics
B-102
Results from a First-In-Human Study with BNZ-1, a Novel, Selective Inhibitor of IL-2, IL-9, and IL-15 at the
Common Gamma-Chain Receptor, in Clinical Development for the Treatment of HAM/TSP and
T-Cell Malignancies
2:05
Michael Caligiuri, MD, Ohio State University
Fc Bridged Cellular Cytotoxicity (FcBCC): a novel mechanism by which NK cells recognize HSV
B-103
2:25
Roberto Accolla, MD, PhD, University of Insubria
Cancer Vaccine: Tumor Immunology meets....Immunology
B-104
2:50
Luigi Buonaguro, MD, National Cancer Institute Fondazione Pascale
Discovery to first-in-man studies of a multi-peptide-based hepatocellular carcinoma vaccine - HEPAVAC
B-105
3:15
Early Stage Investigator: Hua Cheng, PhD, Institute of Human Virology
Development of anti-tumor cytotoxic lymphocytes using engineered human primary blood dendritic cells
B-106
Coffee Break, 3:40 PM - 3:55 PM Grand Prefunction
Session C:
Preventative and Therapeutic Vaccines
Grand Ballroom
Chairpersons and Discussants:
William Blattner, MD, Salt Run Global
George Lewis, PhD, Institute of Human Virology
C-101
3:55
John Moore, PhD, Weill Cornell Medicine
Production and properties of SOSIP trimers
4:20
Ronald Desrosiers, PhD, University of Miami
Long-term delivery of antiviral monoclonal antibodies
4:45
William Schief, PhD, Scripps Research Institute
Germline-targeting vaccine design for HIV
C-103
5:10
Lai-Xi Wang, PhD, University of Maryland College Park
Defining the glycopeptide epitopes of broadly neutralizing antibodies for HIV vaccine design
C-104
19th Annual International Meeting of the Institute of Human Virology
C-102
18
Speaker Schedule
5:35
Paolo Lusso, MD, PhD, National Institute of Allergy and Infectious Diseases
Quaternary Configuration of the HIV-1 Receptor-Binding Site
6:00
Early stage: Mohammad Sajadi, MD, Institute of Human Virology
Pan-neutralizing antibodies derived from human plasma targeting a new epitope
Opening Reception 6:30 PM - 8:30 PM Grand Prefunction
C-105
C-106
Tuesday, October 24, 2017
Session D:
Immunology and Viral Pathogenesis
Grand Ballroom
Chairpersons and Discussants:
Warner Greene, MD, PhD, Gladstone Institutes
Anthony Devico, PhD, Institute of Human Virology
D-101
8:30
Thomas Lehner, MD, Kings College London
The role of stress agents in immunization against HIV-1 infection and eliciting CD4+ memory stem cells
8:55
Guido Silvestri, MD, Emory University
D-102
Immune-based interventions for HIV cure: lessons from NHP models
9:20
John Mellors, MD, University of Pittsburgh
HIV Cure: Time to Rethink the “Shock and Kill” Strategy?
D-103
9:45
Mathias Lichterfeld, MD, Ragon Institute
Clonal proliferation of CD4 T cells encoding intact HIV-1
D-104
Coffee Break, 10:10 AM - 10:25 AM Grand Prefunction
10:25
Guido Poli, MD, Vita-Salute San Raffaele University
Reversible HIV-1 Latency Induced in Primary Human Monocyte-Derived Macrophages by Repeated
M1 Polarization
D-105
10:50
Eric Verdin, MD, The Buck Institute
Chromatin Functional States Correlate with the Reversal of Latently HIV-1 Infected Primary CD4+ T cells
D-106
11:15
Richard Wyatt, PhD, Scripps Research Institute
HIV Env trimer immunogen design and modification to elicit neutralizing antibodies
D-107
11:40
Early Stage Investigator: Keith Reeves, PhD, Harvard Medical School
D-108
Innate-primed alternative signaling pathways enhance functional mucosal mobilization of memory-like NK cells
in HIV/SIV infection
Lunch Break, 12:05 PM - 1:20 PM
19th Annual International Meeting of the Institute of Human Virology
19
Speaker Schedule
Session D:
Immunology and Viral Pathogenesis (continued)
Grand Ballroom
Chairpersons and Discussants:
Anders Vahlne, MD, PhD, Karolinska Institute
Alan Schmaljohn, PhD, University of Maryland School of Medicine
1:20
Warner Greene, MD, PhD, Gladstone Institutes
D-109
Defining a Potent New Class of Latency Reversing Agents Devoid of Toxicity and Detrimental Cell Activation
That Enhance CTL/NK Cell Killing
1:45
Howard Gendelman, MD, University of Nebraska Medical Center
D-110
Combination of CRISPR-Cas9 and Long Acting Slow Effective Release Antiretroviral Therapy Eliminates HIV-1
Infection in Humanized Mice
2:10
Savita Pahwa, MD, University of Miami
D-111
Influence of age on immune response to influenza vaccination in virologically suppressed HIV infected persons
Session E:
Public Health Science and Responses - From Local to Global
Grand Ballroom
Chairpersons and Discussants:
Erica Ollmann Saphire, PhD, Scripps Research Institute
Kathleen Neuzil, MD, MPH, Center for Vaccine Development, Institute of Global Health,
University of Maryland School of Medicine
2:35
Boris Lushniak, MD, MPHD, University of Maryland College Park School of Public Health
Infectious Disease - Public Health Response
E-101
3:00
The Honourable Isaac Adewole, MBBS(Ib), FWACS, FMCOG, FAS, FRCOG, DSc(Hons),
The Federal Minister of Health of Nigeria
E-102
Policy, Practice and Science in Nigeria: Implementing Evidence Based Research for HIV programming in Nigeria
3:25
Kashef Ijaz, MD, MPh, Centers for Disease Control and Prevention
E-103
Global Health Security – Why is it important?
3:50
Early Stage Investigator: Gytis Dudas, PhD, Fred Hutchinson Cancer Research Center
Virus genomes reveal factors that spread and sustain the Ebola epidemic
E-104
Coffee Break, 4:15 PM - 4:30 PM Grand Prefunction
19th Annual International Meeting of the Institute of Human Virology
20
Speaker Schedule
Session F:
Cancer and Stem Cells
Grand Ballroom
Chairpersons and Discussants:
Leonid Margolis, PhD, National Institute of Child Health and Human Development
Guido Poli, MD, Vita-Salute San Raffaele University
4:30
Stavroula Kousteni, PhD, Columbia University
Regulation of Leukemogenesis by the Bone Marrow Niche
F-101
4:55
Heinrich Jasper, PhD, Buck Institute for Research on Aging
Inflammation and immune modulation: tackling age-related stem cell dysfunction
F-102
5:20
Ulrich G. Steidl, MD, PhD, Albert Einstein College of Medicine
Targeting Aberrant Transcription in Pre-Cancerous Stem Cells
F-103
5:45
Early Stage Investigator: Chozha Rathinam, PhD, Institute of Human Virology
F-104
Decoding the regulatory networks of Leukemic Stem Cells through Ubiquitylation
Poster Session 6:10 PM - 8:30 PM Grand Prefunction
Wednesday, October 25, 2017
Session G:
Infectious Agents and Cancer
Grand Ballroom
Chairpersons and Discussants:
Franco Buonaguro, MD, Instituto Nazionale Tumori “Fondazione Pascale”
Kevin Cullen, MD, University of Maryland School of Medicine
8:30
Massimo Tommasino, PhD, International Agency for Research on Cancer
G-101
Human papillomaviruses and carcinogenesis: well-established and novel models
8:55
Sam Mbulaiteye, MBChB, MPhil, MMed, National Cancer Institute
G-102
Endemic Burkitt lymphoma and infectious agents in cancer: new results from the EMBLEM study implicating
falciparum malaria and Epstein-Barr virus in causation
9:20
Charles Bangham, ScD, FMedSci, Imperial College London
Regulation of HTLV-1 proviral latency
9:45
Eric Sundberg, PhD, Institute of Human Virology
G-104
The Helicobacter pylori adhesin protein HopQ exploits the dimer interface of human CEACAMs for oncoprotein
translocation
G-103
Coffee Break, 10:10 AM - 10:25 AM Grand Prefunction
19th Annual International Meeting of the Institute of Human Virology
21
Speaker Schedule
G-105
10:25
Giorgio Trinchieri, MD, National Cancer Institute
Role of Microbiome in Cancer
10:50
Cynthia Sears, MD, Johns Hopkins University
G-106
Carcinogenic Potential of Bacterial Biofilms
11:15
Cornelia Trimble, MD, Johns Hopkins University
HPV vaccines: where are we now, and where are we going?
G-107
11:40
Nicolas Wentzensen, MD, PhD, National Cancer Institute
Molecular Etiology of Cervical Cancer, HPV and gene methylation
G-108
Lunch Break, 12:05 PM - 2:15 PM
Session H:
Lifetime Achievement Award Mini-Symposium
Grand Ballroom
Chairpersons and Discussants:
Robert Gallo, MD, Institute of Human Virology
Manfred Dierich, MD, Medical University Innsbruck
2:15
Robert C. Gallo, MD, Institute of Human Virology
Introduction to Lifetime Achievement Awards
2:20
Mary Klotman, MD, Duke University
H-101
Speaking in honor of Peter Palese: Potential of Integrase-defective Lentiviral Vectors for HIV vaccine delivery
Bernard Roizman, ScD, University of Chicago
H-102
Speaking in honor of Peter Palese: The détente between herpes simplex viruses and their human host
2:50
3:20
Peter Palese, MD, Mount Sinai Medical School
Reinhard Kurth Memorial Lecture: Towards a Universal Influenza Virus Vaccine
H-103
Coffee Break, 4:00 PM - 4:15 PM Grand Prefunction
Session H:
Lifetime Achievement Award Mini-Symposium (continued)
Chairpersons and Discussants:
Robert Redfield, MD, Institute of Human Virology
The Honourable Isaac Adewole, MBBS(Ib), FWACS, FMCOG, FAS, FRCOG, DSc(Hons), The Federal Minister
of Health of Nigeria
4:15
Thomas Quinn, MD, MSc, NIAID, National Institutes of Health
Speaking in honor of Quarraisha Abdool Karim: The HIV Pandemic in Sub-Saharan Africa: Challenges,
Successes and Leadership
H-104
4:45
John Martin, PhD, Gilead Sciences
Speaking in honor of Salim Abdool Karim: Reflections on the Impact of Nucleotide Antiviral Drugs
H-105
19th Annual International Meeting of the Institute of Human Virology
22
Speaker Schedule
5:15
Anthony Fauci, MD, National Institutes of Health
H-106
Speaking in honor of Salim Abdool Karim and Quarraisha Abdool Karim: Sustained ART-Free HIV Remission:
Obstacles and Opportunities
6:00 - 7:00 PM Gala Reception Grand Prefunction
7:00 PM Lifetime Achievement Awards Dinner Cobalt Ballroom
Thursday, October 26, 2017
Session I:
Advances in Clinical Virology - Cardiovascular and Liver Complications of Viral Infections
Grand Ballroom
Chairpersons and Discussants:
Shyam Kottilil, MD, Institute of Human Virology
Ed Tramont, MD, National Institute of Allergy and Infectious Diseases
8:30
Henry Masur, MD, National Institutes of Health
The Changing Natural History of HIV Infection: From Opportunistic Infections to Inflammation and Chronic
Diseases
I-101
8:55
Robert Weiss, MD, Johns Hopkins University
Probing mechanisms of accelerated coronary atherosclerosis and heart disease in HIV+ people
I-102
9:20
Barry Peters, MBBS, MD, FCRP, Kings College London
The Metabolic Complications of HIV
I-103
9:45
Early Stage Investigator: Shashwatee Bagchi, MD, Institute of Human Virology
Cardiovascular Disease in HIV and Chronic Hepatitis C-Infected Patients
I-104
Coffee Break, 10:10 AM - 10:25 AM Grand Prefunction
10:25
Patrizia Farci, MD, National Institutes of Health
Advances in HCV-Associated Hepatocellular Carcinoma
I-105
10:50
Kenneth Sherman, MD, PhD, University of Cincinnati
Hepatitis Viruses in HIV Infection
I-106
11:15
Angus Dalgleish, MD, FMedSci, St. George’s, University of London
Optimisation of a therapeutic vaccine; the importance of immune modulation
I-107
11:40
Mario Stevenson, PhD, University of Miami Miller School of Medicine
Assessing the contribution of myeloid cells to HIV-1 persistence under ART
I-108
Lunch Break, 12:05 PM - 1:05 PM
19th Annual International Meeting of the Institute of Human Virology
23
Speaker Schedule
Session J:
Special Symposium on Advances in Viral Diagnostics
Grand Ballroom
Chairpersons and Discussants:
Niel Constantine, PhD, Institute of Human Virology
Richard Zhao, PhD, University of Maryland School of Medicine
1:05
Niel Constantine, PhD, Institute of Human Virology
Brief Introduction
1:10
Sheila Peel, MSPH, PhD, U.S. Military HIV Research Program
Special Lecture: Current Challenges in HIV Diagnostics – Time to Revise our Approach?
J-101
J-102
1:50
Mark Manak, PhD, U.S. Military HIV Research Program
Decreased levels of seroreactivity in individuals subjected to antiretrovirals early in acute HIV infection
2:10
Michael Reed, PhD, OraSure Technologies
J-103
Performance and usability of the OraQuick® HIV Self-Test, an oral fluid based HIV self-test, in South Africa
2:30
Kathy Shriver, PhD, Bio-Rad
Geenius HIV 1/2 Supplemental Assay: A Rapid, Reliable and Simple System for the Confirmation and
Differentiation of Antibodies to HIV
J-104
2:50
Kevin Delaney, MPH, PhD, Centers for Disease Control and Prevention
Recent developments in HIV testing in the US: Where we are, what’s new, and where we hope to go
J-105
3:10
Rudy Ippodrino, PhD, Ulisse Biomed
Programmable nucleic acid nanoswitches for the rapid, single-step detection of antibodies in bodily fluids
J-106
3:30
Adjourn
19th Annual International Meeting of the Institute of Human Virology
24
Abstracts Index
A-101 Monsef Benkirane, PhD, University of Montpellier
Understanding for targeting the HIV reservoir
A-102 Guido Ferrari, MD, Duke University
Antibody binding to HIV-1 infected cells as mechanism for treatment of HIV infection
A-103 Mirko Paiardini, PhD, Emory University
Combined immune-based strategies targeting viral persistence in ART-suppressed SIV-infected rhesus macaques
A-104 Douglas Nixon, MD, PhD, George Washington University
Immune Targeting of the Latent Reservoir
A-105 Mary Kearney, PhD, National Cancer Institute
No evidence for HIV Replication in lymph nodes during ART
A-106 Kevin Morris, PhD, Beckman Research Institute of the City of Hope
Eradication of HIV; molecular therapeutic paths to a functional cure
A-107 Jintanat Ananworanich, MD, PhD, US Military HIV Research Program
Early ART and HIV Remission: Experience from the RV254 and related HIV remission studies
A-108 Deanna Kulpa, PhD, Emory University
The Contribution of Memory CD4+ T cell subset phenotype to latency reversal efficiency
A-109 Ya-Chi Ho, MD, PhD, Johns Hopkins University
Cytotoxic T lymphocytes shape the landscape of HIV-1 proviruses
B-101 Yutaka Tagaya, MD, PhD, Institute of Human Virology
Shooting Many Cytokines with a Single Stone - a Novel Anti-cytokine Technology with Broad Clinical Application
B-102 Paul Frohna, MD, PhD, PharmD, Bioniz Therapeutics
Results from a First-In-Human Study with BNZ-1, a Novel, Selective Inhibitor of IL-2, IL-9, and IL-15 at the
Common Gamma-Chain Receptor, in Clinical Development for the Treatment of HAM/TSP and T-Cell
Malignancies
B-103 Michael Caligiuri, MD, Ohio State University
Fc Bridged Cellular Cytotoxicity (FcBCC): a novel mechanism by which NK cells recognize HSV
B-104 Roberto Accolla, MD, PhD, University of Insubria
Cancer Vaccine: Tumor Immunology meets...Immunology
B-105 Luigi Buonaguro, MD, National Cancer Institute “Fondazione Pascale”
Discovery to first-in-man studies of a multi-peptide-based hepatocellular carcinoma vaccine - HEPAVAC
B-106 Hua Cheng, PhD, Institute of Human Virology
Development of anti-tumor cytotoxic lymphocytes using engineered human primary blood dendritic cells
C-101 John Moore, PhD, Weill Cornell Medicine
Production and properties of SOSIP trimers
19th Annual International Meeting of the Institute of Human Virology
25
Abstract Index
C-102 Ronald Desrosiers, PhD, University of Miami
Long-term Delivery of Antiviral Monoclonal Antibodies
C-103 William Schief, PhD, Scripps Research Institute
Germline-targeting vaccine design for HIV
C-104 Lai-Xi Wang, PhD, University of Maryland College Park
Defining the glycopeptide epitopes of broadly neutralizing antibodies for HIV vaccine design
C-105 Paolo Lusso, MD, PhD, National Institute of Allergy and Infectious Diseases
Quaternary Configuration of the HIV-1 Receptor-Binding Site
C-106 Mohammad Sajadi, MD, Institute of Human Virology
Pan-neutralizing antibodies derived from human plasma targeting a new epitope
D-101 Thomas Lehner, MD, Kings College London
The role of stress agents in immunization against HIV-1 infection and eliciting CD4+ memory stem cells
D-102 Guido Silvestri, MD, Emory University
Immune-based interventions for HIV cure: lessons from NHP models
D-103 John Mellors, MD, University of Pittsburgh
HIV Cure: Time to Rethink the “Shock and Kill” Strategy?
D-104 Mathias Lichterfeld, MD, Ragon Institute
Clonal proliferation of CD4 T cells encoding intact HIV-1
D-105 Guido Poli, MD, Vita-Salute San Raffaele University
Reversible HIV-1 Latency Induced in Primary Human Monocyte-Derived Macrophages by Repeated M1
Polarization
D-106 Eric Verdin, MD, The Buck Institute
Chromatin Functional States Correlate with the Reversal of Latently HIV-1 Infected Primary CD4+ T cells
D-107 Richard Wyatt, PhD, Scripps Researchg Institute
HIV Env trimer immunogen design and modification to elicit neutralizing antibodies
D-108 R. Keith Reeves, PhD, Harvard Medical School
Innate-primed alternative signaling pathways enhance functional mucosal mobilization of memory-like NK cells
in HIV/SIV infection
D-109 Warner Greene, MD, PhD, Gladstone Institutes
Defining a Potent New Class of Latency Reversing Agents Devoid of Toxicity and Detrimental Cell Activation
That Enhance CTL/NK Cell Killing
D-110 Howard Gendelman, MD, University of Nebraska Medical Center
Synergism between CRISPR/Cas9 and LASER ART leads to elimination of HIV-1 with no rebound
in Humanized Mice
19th Annual International Meeting of the Institute of Human Virology
26
Abstract Index
D-111
Savita Pahwa, MD, University of Miami
Influence of age on immune response to influenza vaccination in virologically suppressed HIV infected persons
E-101 Boris Lushniak, MD, MPHD, University of Maryland College Park School of Public Health
Infectious Disease - Public Health Response
E-102 The Honourable Isaac Adewole, MBBS(Ib), FWACS, FMCOG, FAS, FRCOG, DSc(Hons), The Federal Minister
of Health of Nigeria
Policy, Practice and Science in Nigeria: Implementing Evidence Based Research for HIV programming in Nigeria
E-103 Keshef Ijaz, MD, MPh, Centers for Disease Control and Prevention
Global Health Security – Why is it important?
E-104 Gytis Dudas, PhD, Fred Hutchinson Cancer Research Center
Virus genomes reveal factors that spread and sustained the Ebola epidemic
F-101 Stavroula Kousteni, PhD, Columbia University
Regulation of Leukemogenesis by the Bone Marrow Niche
F-102 Heinrich Jasper, PhD, Buck Institute for Research on Aging
Inflammation and immune modulation: tackling age-related stem cell dysfunction
F-103 Ulrich Steidl, MD, PhD, Albert Einstein College of Medicine
Targeting Aberrant Transcription in Pre-Cancerous Stem Cells
F-104 Chozha Rathinam, PhD, Institute of Human Virology
Decoding the regulatory networks of Leukemic Stem Cells through Ubiquitylation
G-101 Massimo Tommasino, PhD, International Agency for Research on Cancer
Human papillomaviruses and carcinogenesis: well-established and novel models
G-102 Sam Mbulaiteye, MBChB, MPhil, MMed, National Cancer Institute
Endemic Burkitt lymphoma and infectious agents in cancer: new results from the EMBLEM study implicating
falciparum malaria and Epstein-Barr virus in causation
G-103 Charles Bangham, ScD, FMedSci, Imperial College London
Regulation of HTLV-1 proviral latency
G-104 Eric Sundberg, PhD, Institute of Human Virology
The Helicobacter pylori adhesin protein HopQ exploits the dimer interface of human CEACAMs for
oncoprotein translocation
G-105 Giorgio Trinchieri, MD, National Cancer Institute
Role of Microbiome in Cancer
G-106 Cynthia Sears, MD, Johns Hopkins University
Carcinogenic Potential of Bacterial Biofilms
19th Annual International Meeting of the Institute of Human Virology
27
Abstract Index
G-107 Cornelia Trimble, MD, Johns Hopkins Medical Institute
HPV vaccines: where are we now, and where are we going?
G-108 Nicolas Wentzensen, MD, PhD, National Cancer Institute
Molecular Etiology of Cervical Cancer, HPV and gene methylation
H-101 Mary Klotman, MD, Duke University
Potential of Integrase-defective Lentiviral Vectors for HIV vaccine delivery
H-102 Bernard Roizman, ScD, University of Chicago
The détente between herpes simplex viruses and their human host
H-103 Peter Palese, MD, Mount Sinai Medical School
Towards a Universal Influenza Virus Vaccine
H-104 Thomas Quinn, MD, MSc, NIAID, National Institutes of Health
The HIV Pandemic in Sub-Saharan Africa: Challenges, Successes and Leadership
H-105 John Martin, PhD, Gilead Sciencies
Reflections on the Impact of Nucleotide Antiviral Drugs
H-106 Anthony Fauci, MD, NIAID, National Institutes of Health
Sustained ART-Free HIV Remission: Obstacles and Opportunities
I-101
Henry Masur, MD, National Institutes of Health
The Changing Natural History of HIV Infection: From Opportunistic Infections to Inflammation and
Chronic Diseases
I-102
Robert Weiss, MD, Johns Hopkins University
Probing mechanisms of accelerated coronary atherosclerosis and heart disease in HIV+ people
I-103
Barry Peters, MBBS, MD, FCRP, King’s College London
The Metabolic Complications of HIV
I-104
Shashwatee Bagchi, MD, Institute of Human Virology
Cardiovascular Disease in HIV and Chronic Hepatitis C-Infected Patients
I-105
Patrizia Farci, MD, National Institutes of Health
Advances in HCV-Associated Hepatocellular Carcinoma
I-106
Kenneth Sherman, MD, PhD, University of Cincinnati
Hepatitis Viruses in HIV Infection
I-107
Angus Dalgleish, MD, FMedSci, St. George’s, University of London
Optimisation of a therapeutic vaccine; the importance of immune modulation
I-108
Mario Stevenson, PhD, University of Miami Miller School of Medicine
Assessing the contribution of myeloid cells to HIV-1 persistence under ART
19th Annual International Meeting of the Institute of Human Virology
28
Abstract Index
J-101
Sheila Peel, MSPH, PhD, US Military HIV Research Program
Current Challenges in HIV Diagnostics – Time to Revise our Approach?
J-102
Mark Manak, PhD, US Military HIV Research Program
Decreased levels of seroreactivity in individuals subjected to antiretroviral therapy early in acute HIV infection
J-103
Michael Reed, PhD, OraSure Technologies
Performance and usability of the OraQuick® HIV Self-Test, an oral fluid based HIV self-test, in South Africa
J-104
Kathleen Shriver, PhD, Bio-Rad
Geenius HIV 1/2 Supplemental Assay: A Rapid, Reliable and Simple System for the Confirmation and
Differentiation of Antibodies to HIV
J-105
Kevin Delaney, MPH, PhD, Centers for Disease Control and Prevention
Recent developments in HIV testing in the US: Where we are, what’s new, and where we hope to go
J-106
Rudy Ippodrino, PhD, Ulisse Biomed
Programmable nucleic acid nanoswitches for the rapid, single-step detection of antibodies in bodily fluids
19th Annual International Meeting of the Institute of Human Virology
29
Poster Index
P-A1
Mudit Tyagi, PhD, The George Washington University
CBF-1 promotes the establishment and maintenance of HIV latency by recruiting Polycomb repressive
complexes, PRC1 and PRC2, at HIV LTR
P-A2
Takashi Okamoto, MD, PhD, Nagoya City University
Development of anti-Tat compound: MD Simulation of the Tat/Cyclin T1/CDK9 Complex Revealing the Hidden
Catalytic Cavity within the CDK9 Molecule Upon Tat Binding
P-A3
Richard Apps, PhD, The George Washington University
Limitations of CD32a expression as a marker of the HIV latent reservoir
P-A4
Christopher Woldstad, PhD, University of Nebraska Medical Center
Multimodal theranostic tests for antiretroviral drug delivery
P-A5
Robert Furler, PhD, The George Washington University
HIV Infected Cells Have Depolarized Membrane Potentials and Increased Intracellular Calcium Levels
P-A6
Jianshi Yu, PhD, University of Maryland School of Pharmacy
A Possible Role for Retinoic Acid in the Functional Cure of SIV Infections in ART/α4β7 mAb-treated Monkeys
P-A7
Tracy Evans-Gilbert, MD,MPH,CTropMed®, Jamaica Perinatal and Paediatric HIV/AIDS Programme, Cornwall
Regional Hospital
HIV disease history in perinatally HIV infected adolescents with interrupted care
P-A8
Hang Su, BS, University of Nebraska Medical Center
Establishing tissue reservoirs for the human immunodeficiency virus in humanized mice
P-A9
Mary Banoub, BS, University of Nebraska Medical Center
Transformation of Darunavir into a long acting Nanoformulated Prodrug
P-A10 Elizabeth Anderson, B.Sci, HIV Dynamics and Replication Program, National Cancer Institute
Accumulation and persistence of deleted HIV proviruses following prolonged ART
P-A12 Camille Lange, MSc, PhD, National Cancer Institute, National Institute of Health, Frederick, MD
Discordant HIV Populations with Discordant V3 Tropism in CSF and Plasma: Implications for Establishing HIV
Reservoirs
P-C1
Peter Smith, PhD, St. Georges University of London
Development of HIV-1 Immunotherapy with Vacc4x and Vacc-C5
P-C2
Yongjun Sui, PhD, National Cancer Institute
Protection against or delay of intrarectal SHIV acquisition by mucosal vaccines in the absence of Env-antibody
responses
P-C3
Camila Guzman, BS, Institute of Human Virology
A Modified Vaccinia Ankara vector expressing Lassa virus-like particles (MVA-LasVLP) protects mice from lethal
challenge with a Lassa-Mopeia reassortant virus
19th Annual International Meeting of the Institute of Human Virology
30
Poster Index
P-C4
Walther Mothes, PhD, Yale University
Associating HIV-1 Env Trimer Structures with Functional Env Conformational States by smFRET Analysis
P-C5
Ursula Dietrich, PhD student, Georg-Speyer-Haus
Broadly neutralizing nanobodies selected from dromedary immune libraries with subtype C SOSIP Env
glycoproteins: optimization and preclinical development
P-D1
John Williams, PhD, University of Pittsburgh
Differential pathogenesis of human metapneumovirus clinical isolates in C57BL/6 mice
P-D2
Roberto Accolla, PhD, University of Insubria
TRIM22 binds to CIITA and sequesters it into nuclear bodies containing TRIM19/PML and Cyclin T1.
Implications for HIV-1 infection
P-D4
Orrianne Morrison, PhD, Food and Drug Administration
HIV-1 Env Trimer Conformational Implications of Peptide Fusion Inhibitor Resistance
P-D6
Jibreel Jumare, MBBS, PhD, University of Maryland at Baltimore
Elevated Plasma HIV RNA level is associated with impaired neurocognitive function among HIV-1 infected
patients in Nigeria
P-D8
Chiara Orlandi, PhD, Institute of Human Virology
New insights on the human anti-HIV-1 Env antibody-mediated cell cytotoxicity (ADCC) against HIV-1 virus:
Allosteric regulation of FcRs binding upon antigen engagement
P-E1
Richard Zhao, PhD, Institute of Human Virology
A novel way to test and discover new inhibitors against drug resistant HIV-1 proteases
P-E2
Nadia Sam-Agudu, MPH, International Research Center of Excellence, Institute of Human Virology Nigeria
The Impact of Structured Mentor Mother Support on Retention During the First 12 Months Postpartum among
HIV Positive Women in Rural Nigeria
P-E3
Niel Constantine, PhD, Institute of Human Virology
Assessment of internationally-available HIV test kits for their suitability to meet manufacturers’ claims
P-E4
Nadia Sam-Agudu, MBBS, MPH, PhD, Institute of Human Virology
The MoMent Study: Correlates of Viral Suppression at 6 months Postpartum among HIV-Positive Women in
Rural Nigeria
P-E5
Nicaise Ndembi, MPhiL, PhD, Institute of Human Virology Nigeria
Whole Genome Deep Sequencing of HIV Reveals Extensive Multi-class Drug Resistance in Nigerian Patients
Failing First-line Antiretroviral Therapy
P-E6
Janet Itelima, PhD, University of Jos, Nigeria
Hepatitis B and Hepatitis C viral infections amoung pregnant women in some Nigerian major Cities: A Review
P-G2
Juan Zapata, Msc, PhD, Institute of Human Virology
Development of engineered T-cell immmunotherapy for treating Adult-T cell leukemia caused by HTLV-1
19th Annual International Meeting of the Institute of Human Virology
31
Poster Index
P-G3
Terry-Elinor Reid, PhD, Institute of Human Virology
Unravelling the Mechanism of Action of HLBT-100 Molecule against Adult T-Cell Leukemia
P-G4
Makar Tapas, PhD, Department of Neurology, University of Maryland
Molecular characterization of hippocampal changes in an animal model of HIV associated Primary Central
Nervous System Lymphoma
P-G5
Franco Maria Buonaguro and AnnaLucia Tornesella, Istituto Nazionale Tumori – IRCCS Fondazione Pascale
Differential immune response to HCV peptides as cancer-progression biomarkers of HCV-infections
P-I1
Alonso Heredia, PhD, Instiute of Human Virology
Monotherapy with Integrase Inhibitors Does Not Maintain Viral Suppression in Humanized Mice with Chronic
HIV Infection
P-J1
Paul Lambotte, PhD, Chembio Diagnostic Systems, Inc.
Meta-analysis of laboratory and field studies of the DPP® HIV Syphilis Assay, a single-use, rapid combination
test for the detection of HIV and syphilis antibodies at the Point of Care
19th Annual International Meeting of the Institute of Human Virology
32
Speaker Abstracts
A-101
Understanding for targeting the HIV reservoir
Monsef Benkirane, PhD, University of Montpellier
HIV infection is suppressed but not cured by Anti-Retroviral Therapy (ART).
This is due to the establishment of a viral reservoir that is responsible for
the persistence of low levels of plasma viremia in patients under ART. Viral
rebound is observed immediately after ART interruption. HIV persistence
may arise from ongoing residual virus replication and/or from latentlyinfected cells in which long-lived resting memory CD4+ T cells harbouring
transcriptionally silent provirus represent the largest pool in the blood.
Addressing the source of HIV persistence is required to achieve a cure for
HIV. We will discuss our recent data showing that CD32a is marker of HIV
persistent CD4 T cell.
A-102
Antibody binding to HIV-1 infected cells as mechanism for
treatment of HIV infection
Guido Ferrari, MD, Duke University; Jeffrey Nordstrom, PhD,
Macrogenics; Scott Koenig, MD, Macrogenics; Julia Sung, MD, University
of North Carolina - Chapel Hill; David Margolis, MD, University of North
Carolina - Chapel Hill; Barton Haynes MD, Duke University Medical Center
Anti-HIV antibody function is not limited to capturing free virus but also
includes the recognition of HIV-1 infected cells. Both neutralizing and nonneutralizing antibody responses recognizing HIV-1 envelope epitopes
expressed on the cellular membrane can recruit Fc receptor-bearing cells
and direct their cytotoxic activity against the infected cells. We can leverage
on our ability to use monoclonal antibody and monoclonal antibody-based
molecules to protect from and treat HIV-1 infection.
A-103
Combined immune-based strategies targeting viral persistence in
ART-suppressed SIV-infected rhesus macaques
Mirko Paiardini, PhD, Emory University
Antiretroviral therapy (ART) suppresses viral replication in HIV-infected
individuals, but does not eliminate an extremely durable reservoir of
latently infected cells that is established early after infection. Understanding
the phenotype and location of latently infected cells represents a critical
challenge in designing a cure for HIV; furthermore, many HIV-infected
individuals given ART exhibit residual inflammation, which may contribute to
virus persistence. This presentation will discuss immune based strategies
targeting HIV persistence developed over the past several years using the
model of SIV infection in rhesus macaques. Recent work identified PD1+ follicular helper CD4+ T-cells as an important cellular compartment for
viral persistence. We have described that CTLA-4+PD-1- memory CD4+
T-cells, which share phenotypic markers with regulatory T-cells and localize
outside the B-cell follicle of the lymph nodes, are significantly enriched in
SIV-DNA; contain robust levels of replication-competent virus; and increase
their contribution to the SIV reservoir with prolonged ART. Finally, we
showed that Interleukin-21 and IFN-alpha administration in ART-treated,
SIV-infected RMs reduces residual immune activation during ART and
temporally delay viral rebound after ART treatment interruption.
These recent advancements highlight the complexity and diversity of the
mechanisms and T-cell populations that can contribute to the residual
reservoirs of virally infected cells. Developing a range of different
interventions to target individual components of viral reservoirs represent
both a formidable challenge and an exciting opportunity for the years to
come.
A-104
Immune Targeting of the Latent Reservoir
Douglas F. Nixon, MD, PhD, The George Washington University
HIV is a human infectious exogenous retrovirus which is thought to have
infected human populations from cross primate species transmissions.
Within our genomes are remnants of past infectious retroviruses which
have endogenized, called “endogenous retroviruses” (ERVs). We have
previously shown that human ERVs (HERVs) can be transcribed in an HIV
infected cell, but had not identified which sub-family or unique HERV was
transcribed. Using a novel computational pipeline, Telescope, we have
identified which HERVs are expressed in HIV infected cells.
We have received funding for a Martin Delaney Collaboratory grant called
“Believe” in which we research towards HIV eradication based upon
enhanced targeted cell therapy. As part of this program, we have investigated
whether we can identify markers which uniquely identify the latent reservoir.
New data will be presented on two putative markers, CD32a and IFITM1.
Funding: The NIH Martin Delaney Collaboratory, “Believe”, NIAID UM1
award AI126617, co-funded by NIDA/NINDS/NIMH/NIAID; and grants NCI
CA 206488; NIAID AI 76059.
19th Annual International Meeting of the Institute of Human Virology
33
Speaker Abstracts
A-105
NO EVIDENCE FOR HIV REPLICATION IN LYMPH NODES
DURING ART
William McManus, BS, National Cancer Institute - Frederick; Jonathan
Spindler, BS, National Cancer Institute - Frederick; Michael Bale, BS,
National Cancer Institute - Frederick; Ann Wiegand, MS, National Cancer
Institute - Frederick; Andrew Musick, BS, National Cancer Institute Frederick; Xiaolin Wu, PhD, Liedos Biomedical; David Wells, BS, Liedos
Biomedical; Stephen Hughes, PhD, National Cancer Institute - Frederick;
Mary Kearney, PhD, National Cancer Institute – Frederick; et al
To better understand the mechanisms of HIV persistence and to further
investigate the question on ongoing viral replication in lymph nodes, we
characterized HIV proviral populations, their levels of expression, and their
sites of host integration in paired lymph node mononuclear cells (LNMC)
and peripheral blood mononuclear cells (PBMC) collected after long-term
ART and compared to them to the HIV populations prior to ART initiation.
Three donors initiated ART in chronic infection and had viremia suppressed
for 4-12 years; one donor initiated ART in acute infection and had viremia
suppressed for 18 years. Proviral populations and expression were
characterized by single-cell analyses and expanded clones were identified
using the integration sites assay (ISA). Populations were compared
phylogenetically, using a test for panmixia (well-mixed or divergent), by
determining the fraction of expressing vs. latent proviruses, determining
the levels of expression in single cells with actively-transcribing proviruses,
and by comparing the expanded clonal populations. Proviruses in LNMC
and PBMC were well mixed and were not significantly divergence from the
pre-ART populations. The fraction of proviruses that were latent vs. actively
transcribing during ART were not different (5-8% in the PBMC and 2-20%
in the LNMC) (p=0.4). The levels of expression in actively-infected cells
were low in both compartments, typical of suppression of viral replication.
The same clonal populations were detected in PBMC and LNMC (p=0.8).
These findings are not consistent with continued viral replication during
ART in either the peripheral blood or the lymph nodes. Our results also
suggest that infected cells migrate freely between the peripheral blood and
the lymph nodes and demonstrate that the HIV reservoir is long-lived and
proliferating populations of cells that were infected prior to initiating ART.
A-106
Eradication of HIV; molecular therapeutic paths to a functional
cure
Kevin Morris, PhD, The Beckman Research Institute at the City of Hope
The eradication of functional replicative HIV is within our collective grasp.
Several emerging modes of targeted therapeutics, ranging from chimeric
antigen receptor (CAR) T-cell targeted killing of virus infected cells to
targeted genomic editing and/or excision of latent provirus, may prove
promising with regards to eradicating the spread of functional HIV. We have
recently developed two distinct molecular approaches that may facilitate
the eradication of HIV. The first approach is the systemic activation of HIV
followed by a polyvalent “swarm” of HIV-gp120 bi-specific CAR CD4+/
CD8+ T-cells. Systemic activation of latent HIV can occur by removing
anti-retroviral therapy (ART) and/or by targeted activation of provirus. To
activate provirus we developed a biologically deliverable recombinant zinc
finger protein activator (ZFP-362-VPR), that functionally activates HIV in
an LTR directed and specific manner. Notably, ZFP-362-VPR can be used
in combination with a swarm of differentially HIV-gp120 targeted bi-specific
CAR CD4+/CD8+ T-cells, to specifically kill virus infected cells. A second
approach to eradicate HIV is the targeted genomic editing and/or excision
of integrated virus. The approach we describe here builds on biologically
deliverable recombinant protein technologies to direct the excision and/or
targeted mutations to loci in the provirus, ultimately rendering the virus nonreplicative. Collectively, these emerging molecular therapeutic strategies
juxtaposed with current antiretroviral therapy may one day result in the
eradication of HIV.
A-107
Early ART and HIV Remission: Experience from the RV254 and
related HIV remission studies
Jintanat Ananworanich, MD, PhD, US Military HIV Research Program
(MHRP)
Early antiretroviral therapy (ART) is an important step in the path towards
an HIV cure. Acute HIV infection studies have demonstrated the ability
to identify and treat very early infection in adults, resulting in significantly
smaller HIV reservoirs, preserved immune functions with little viral escape
to immune pressure. These results have been illustrated in the ongoing
RV254 acute HIV infection study in Thailand that has enrolled close to 500
individuals. It has served as a platform to learn the effects of early ART and
recruit participants for HIV remission trials.
Despite the favorable qualities, the majority of early treated individuals do
not achieve HIV remission. Indeed this has been the experience to date in
the RV254 HIV remission trials, which include three completed studies and
one ongoing, with others in development. The interventions have included
latency-reversing agent, broadly neutralizing antibody and therapeutic HIV
vaccine. However, there are important lessons to be learned that would
inform the design of future HIV remission trials.
There continues to be exciting new development of immune-based
interventions. It is highly likely that combination therapies that can generate
persistent and effective immune responses to control HIV will be required
for a durable ART-free sustained remission of HIV.
19th Annual International Meeting of the Institute of Human Virology
34
Speaker Abstracts
A-108
B-101
The Contribution of Memory CD4+ T cell subset phenotype to
latency reversal efficiency
Shooting Many Cytokines with a Single Stone - a Novel Anticytokine Technology with Broad Clinical Application.
Deanna Kulpa, PhD, Emory University; Aarthi Talla, PhD, Case Western
Reserve University; Susan Ribeiro, PhD, Case Western Reserve
University; Richard Barnard, PhD, Merck & Co., Inc.; Daria Hazuda, PhD,
Merck & Co., Inc.; Nicolas Chomont, PhD, University of Montreal; Rafick
Sekaly, PhD, Case Western Reserve University
Yutaka Tagaya, MD, PhD, Institute of Human Virology; Juan Zapata,
PhD, Institute of Human Virology; Xiaorong Wu, BS, Institute of Human
Virology; Asjad Basheer, PhD, BIONIZ; Paul Frohna, MD, PhD, BIONIZ;
Nazli Azimi, PhD, BIONIZ
The latent HIV reservoir persists in individuals on ART predominantly in
memory CD4+ T cells, a heterogeneous population comprised of central
memory (CM), transitional memory (TM) and effector memory (EM)
subsets. Current HIV eradication strategies that aim to reverse latency in
this heterogeneous pool of cells have had limited success. To characterize
HIV latency reversal in all memory CD4+ T cell subsets that contribute to the
HIV reservoir in vivo, we developed LARA (Latency and Reversion Assay),
a primary cell based in vitro model of HIV latency. To identify pathways
associated with latency reversal in each subset, we exposed latently
infected cells from both HIV-infected individuals and LARA to different
classes of latency reversing agents (LRAs). Memory subsets showed
distinct responses that resulted in varying efficiencies to the LRAs tested.
Importantly, the most effective LRAs triggered the differentiation into cells
that expressed an EM phenotype. Transcriptional profiling of CD4+ T cells
from HIV-infected individuals exposed to bryostatin, the LRA that showed
the highest latency reversal, identified several EM specific pathways that
were significantly upregulated in both the CM and EM subsets, including
genes encoding for cytokines and effector molecules such as IFN-γ, IL2, IL-4, and TNF. Together, these results support LRA exposure triggering
differentiation toward an EM subset phenotype to be linked to higher
latency reversal efficiency. Identification of these pathways is a critical
prerequisite to understand factors that influence latency reversal in vivo
as well as contributing to the most effective design of regimens capable of
comprehensive reactivation of the HIV reservoir in eradication strategies.
A-109
Cytotoxic T lymphocytes shape the landscape of HIV-1 proviruses
gc-cytokines (IL-2, -4, -7, -9, -15, -21) control major immune responses
and lymphocyte development. Their aberrant production drives multiple
human diseases including HTLV-1 associated myelopathy (HAM), T-cell
malignancies, graft-versus-host (GvH) disease and autoimmunity (e.g.,
alopecia areata, Celiac disease, asthma). Importantly, these diseases are
caused by 2 or more cytokines (multi-cytokine diseases) which cannot be
effectively and safely treated by the current anti-cytokine approaches.
To address this issue, we developed a novel class of multi-cytokine
inhibitors (MCI) that are designed as cytokine-mimetic peptides each of
which contains a short motif shared by only the targeted cytokines of the
c family. Our first MCI, BNZ 132-1, selectively inhibits IL-2, -9, -15 without
affecting other c- or non-c-cytokines. BNZ 132-1 efficiently eradicated
LGL leukemic cells without damaging normal cells in the IL-15 leukemic
model we developed, in which a Jak inhibitor (Tofacitinib) showed only
limited efficacy (Nata et al. 2015 J Biol Chem).
In humanized-mice infected with LCMV( a mouse model of cytokine release
syndrome (CRS)), animals treated with BNZ 132-1 survived while all control
animals died. BNZ 132-1 reversed the cytokine storm (IL-6, TNF-, IFN within 5 days, which is likely caused by the over-production of IL-2/
IL-15. Similarly, GvH naturally occurring in humanized mice was effectively
suppressed by the BNZ 132-1 treatment. These results suggest a potential
application of BNZ 132-1 for GvH post transplantation and CRS post
microbial infection, in addition to HAM and T-cell malignancies that we are
targeting in our clinical trials. Thus, our new MCI technology fills in a major
gap of anti-cytokine therapy.
Ya-Chi Ho, MD, PhD, Yale University School of Medicine
Despite antiretroviral therapy, HIV-1 persists in memory CD4+ T cells
creating a barrier to cure. The majority of HIV-1 proviruses are defective due
to packaging signal deletions, APOBEC-mediated G-to-A hypermutations,
large internal deletions, and point mutations. These defective proviruses
are considered clinically irrelevant. Using cells from HIV-1-infected
individuals and reconstructed patient-derived defective proviruses, we
show that defective proviruses can be transcribed to RNAs that are spliced
and translated into viral antigens. Proviruses with defective major splice
donors (MSDs) can activate novel splice sites to produce HIV-1 transcripts.
Cells with HIV-1 proviruses containing defective MSDs can be recognized
by HIV-1-specific cytotoxic T lymphocytes (CTLs). Surprisingly, cells with
proviruses containing lethal mutations upstream of CTL epitopes can also
be recognized by CTLs potentially through aberrant translation. Thus,
expression of defective proviruses complicates the measurement of the
latent reservoir. CTLs may change the landscape of HIV-1 proviruses by
preferential targeting cells with specific types of defective proviruses. The
scope of potential CTL targets may be bigger than the size of the latent
reservoir.
19th Annual International Meeting of the Institute of Human Virology
35
Speaker Abstracts
B-102
Results from a First-In-Human Study with BNZ-1, a Novel,
Selective Inhibitor of IL-2, IL-9, and IL-15 at the Common GammaChain Receptor, in Clinical Development for the Treatment of
HAM/TSP and T-Cell Malignancies
Paul Frohna, MD, PhD, PharmD, Bioniz Therapeutics; Yutaka Tagaya,
MD, PhD, Institute for Human Virology; Anoshie Ratnayake, MD, Bioniz
Therapeutics; Nick Doerr, PhD, Bioniz Therapeutics; Asjad Basheer,
PhD, Bioniz Therapeutics; Laith Al-Mawsawi, PhD, Bioniz Therapeutics;
Woo Jae Kim, PhD, Bioniz Therapeutics; Juan Zapata, PhD, Institute for
Human Virology; et al
Aberrant signaling of IL-2, IL-9, and IL-15, members of the γc cytokine
family, is involved in multiple human diseases (eg, T-cell malignancies,
GvHD, HAM/TSP) that are not effectively or safely treated by the currently
available anti-cytokine approaches.
BNZ-1 is a selective inhibitor of IL-2, IL-9 and IL-15 signaling through
γc, without altering IL‑4, -7, or -21. In this open-label, single-dose study
(NCT03046459), 18 healthy adults (n=3/dose) received a single IV dose of
0.2, 0.4, 0.8, 1.6, 3.2 or 6.4 mg/kg and were monitored for 30 days. BNZ-1
was well-tolerated with a good safety profile (no serious/severe AEs, no
DLTs, and no clinically-significant changes on labs, vital signs or ECGs).
BNZ-1 exposure was dose proportional with an elimination t1/2 of ~5
days, supporting weekly or every other week dosing. PD activity was
characterized by flow cytometry of Tregs (IL-2), NK cells (IL-15) and CD8+
central memory T-cells (Tcm; IL-2 & IL-15) in PBMCs from Days 4, 15 & 31.
Tregs were decreased by 50-60% after doses of 0.4-1.6mg/kg on Day 4 and
by 80-93% at 3.2 and 6.4 mg/kg on Day 15. On Day 4 NK cells decreased
by 20, 40 and 60% at 0.2, 0.4 and 0.8mg/kg, respectively, and plateaued at
70-80% decreases at doses ≥1.6 mg/kg. Tcm were decreased at Day 4 for
the top 3 doses that continued to decline to Day 15 when all doses, except
0.2 mg/kg, showed a mean decrease ranging from 10 to 81% that trended
with dose. Tregs, NK cells, and Tcm returned to/toward baseline by Day
31. CD4+ and CD8+ T-cells, B-cells, and monocytes were unchanged at
all time points.
These clinical data suggest that BNZ-1 is a highly-active, selective
immunomodulator that safely decreases Tregs, NKs and Tcm, while sparing
the major leukocyte populations.
19th Annual International Meeting of the Institute of Human Virology
B-103
Fc Bridged Cellular Cytotoxicity (FcBCC): a novel mechanism by
which NK cells recognize HSV
Hongsheng Dai, PhD; Michael A. Caligiuri, MD*
*The Ohio State University Comprehensive Cancer Center; The James
Cancer Hospital and Solove Research Institute, Columbus Ohio
The initial infection by herpes simplex virus 1 (HSV1) is usually a self-limited
event, but the mechanism of this protection prior to the development of a
primary humoral response is unknown. Individuals deficient in natural killer
(NK) cells can suffer from overwhelming and at times fatal HSV1 infection.
The NK cell surface is naturally coated with IgG that is bound to Fcγ receptor
CD16a. Here we show that human NK cells utilize the Fc portion of IgG
bound to its cell surface to recognize gE, an HSV1-encoded glycoprotein
that also binds the Fc portion of IgG but at a site distinct from that of CD16a.
The Fc-bridge formed between the HSV1-infected cell and the NK cell
results in NK cell activation and spontaneous lysis of the HSV-infected cell
in the absence of HSV-specific antibody. In vivo, the absence of the Fc
portion of human IgG, or of NK cells, results in death from primary HSV1
infection. This mechanism, which we call Fc bridged cellular cytotoxicity
(FcBCC) may be broadly applicable to Fcγ receptor-bearing immune cells
and other pathogens encoding Fc-binding proteins.
B-104
Cancer Vaccine: Tumor Immunology meets...Immunology
Roberto Accolla, MD, PhD, University of Insubria
Although recent therapeutic approaches have revitalized the enthusiasm
of the immunological way to combat cancer, still the comprehension of the
immunity against tumors is largely incomplete. Due to their specific function,
CD8+ T cells with cytolytic activity (CTL) have attracted the attention of most
investigators because CTL are considered the main effectors against tumor
cells. Nevertheless, CTL activity and persistence are largely dependent
on the action of CD4+ T helper cells (TH). Thus establishment of tumorspecific TH cell response is key to the optimal response against cancer. I
will describe emerging new strategies to increase the TH cell recognition
of tumor antigens. In particular, I will present recent data indicating that
tumor cells themselves can act as surrogate antigen presenting cells for
triggering TH response if genetically modified to express the MHC class
II transcriptional activator CIITA. Indeed, injection of CIITA-modified tumor
cells of distinct histological origin and of distinct MHC genetic background
into syngeneic recipients induces strong protective and long-lasting adaptive
immune response that can be transferred to naïve recipients by CD4+ TH
cells. Depletion of dendritic cells does not modify the capacity to reject the
tumor and to acquire immunological memory. These results challenge the
immunological dogma that dendritic cells are the exclusive cells capable of
inducing T cell priming. The strategy of modifying tumor cells with CIITA has
been applied for the production of a novel generation of anti-tumor vaccine
against human hepatocarcinoma that is now in clinical trial.
36
Speaker Abstracts
B-105
Discovery to first-in-man studies of a multi-peptide-based
hepatocellular carcinoma vaccine - HEPAVAC
Luigi Buonaguro, MD, National Cancer Institute “Fondazione Pascale”
Hepatocellular (HCC)/normal adjacent tissue matched samples have
been collected for HLA immunopeptidome analysis. 17 HCC samples
from HLA-A*02+ patients and 15 samples from HLA-A*24+ patients have
been analysed by mass spectrometry. RNA-expression profiles have been
established for 12 HCC samples. HLA-presentation/expression of peptides
on primary HCC samples (as well as mRNA expression) were compared to
normal tissue samples from relevant organs (including heart, brain, lung,
kidney, liver, nerve, skin etc.) present in the Immatics’ database.
A total of 16 peptides have been selected and confirmed for immunogenicity
for the HepaVac vaccine and are currently synthesized according to GMP
standard. Of these, 7 are restricted to HLA-A*02; 5 to HLA-A*24 and 4 to
HLA class II.
A single-arm, first-in-man trial entitled HepaVac-101 is designed to
investigate in patients with very early, early and intermediate stage of
HCC the off-the-shelf multi-peptide-based HCC vaccine (IMA970) plus
the CV8102 adjuvant (RNAdjuvant®) following a single pre-vaccination
infusion of low-dose cyclophosphamide acting as an immunomodulator.
The study drugs are applied without concomitant anti-tumor therapy with
the intention to reduce risk of tumor recurrence/progression in patients who
have received all indicated standard treatments. The primary endpoints are
safety, tolerability, and immunogenicity. Secondary/exploratory endpoints
are additional immunological parameters in blood (e.g. regulatory T-cells,
myeloid-derived suppressor cells, impact of the standard therapy on the
natural immune response), infiltrating T-lymphocytes in tumor tissue,
biomarkers in blood and tissue, disease-free survival/progression-free
survival and overall survival.
B-106
Development of anti-tumor cytotoxic lymphocytes using
engineered human primary blood dendritic cells
Long Wu, Huan Zhang, Yixing Jiang, Robert Gallo, Hua Cheng, Institute
of Human Virology, University of Maryland Baltimore School of Medicine
Dendritic cell (DC)-based immunotherapy has achieved modest clinical
benefits, however, several technical hurdles in DC preparation, activation
and cancer-associated antigen (TAA) delivery limit its broad applications
in cancer therapy. We have developed immortalized and activated human
primary blood dendritic cell (DC) lines, ihv-DCs. Human primary blood
dendritic cells can be immortalized from several cc of peripheral blood
and can be expanded at large quantity at normal cell culture condition.
The ihv-DCs are a subset of CD11c+/CD205+ DCs that persistently
display co-stimulatory molecules and produce interferon gamma. These
DCs are engineered to constitutively express a TAA such as hTERT,
which prime donor-derived T cells to generate antigen-specific CTLs that
induce cytolysis of hTERT-expressing target cells in an HLA-A2-restricted
manner. In addition, the engineered DCs are able to induce simultaneous
production of both anti-cancer CTLs and NK cells from naïve PBMCs. In
NSG mouse model, infusion of the activated CTLs and NK cells that are
generated from naïve PBMCs using the engineered DCs suppress lung
metastasis of human lung cancer cells. Both CTLs and NK cells are found
to infiltrate lung as well as lymphoid tissues, mimicking the in vivo trafficking
patterns of cytotoxic lymphocytes. This new approach should facilitate the
development of cell-based immunotherapy for human lung cancer.
C-101
Production and properties of SOSIP trimers
John Moore, PhD, Weill Cornell Medicine
I will review the progress of our team’s projects to produce GMP-quality
SOSIP trimers in amounts and quantities appropriate for human clinical
trials. The BG505 SOSIP.664 GMP-project has now been completed
successfully, yielding ~5 grams of trimers. A follow-up on effort to make the
germline–bNAb targeting BG505 SOSIP.v4.1-GT1.1 trimer is well underway,
using the same core methodologies. The latter trimer was designed at the
Amsterdam Medical Center and is very similar to one recently described by
Medina-Ramirez et al. in J Exp Med.
This project stems from our NIH-funded HIVRAD grant program and the
translation phase has been supported and managed by the BMGF and
IAVI, reflecting the three funding agencies working together towards a
common goal.
19th Annual International Meeting of the Institute of Human Virology
37
Speaker Abstracts
C-102
Long-term Delivery of Antiviral Monoclonal Antibodies
Ronald Desrosiers, PhD, University of Miami Miller School of Medicine
The current availability of an incredible array of monoclonal antibodies
(mAbs) with potent neutralizing activity against a broad range of HIV1 isolates creates attractive possibilities for treatment and prevention.
An ability to achieve stable levels of protective mAb concentrations
long-term via vector delivery has a number of distinct advantages over
repeated passive administrations. These include cost, issues relating to
adherence, and convenience. Using AAV as vector, we have achieved
stable concentrations of authentic IgG mAb in monkeys in excess of 100 ug
per ml of serum for as long as 3.5 years, the longest we have measured.
In the one SHIV-infected monkey to which we have successfully delivered
two anti-HIV mAbs, viral loads have been impressively suppressed from
11,000 at the time of AAV administration to below the limit of detection for
more than 22 months in the absence any antiviral drug treatment. The
main obstacle hindering development of this approach for use in humans
is anti-drug antibodies, i.e. the development of an antibody response to the
delivered antibody that can severely impair the concentrations of delivered
antibody that can be achieved. These anti-antibodies are predominantly or
exclusively directed to the variable domains, i.e. they are anti-idiotype. This
is especially a problem because the mAbs with potent neutralizing activity
that we want to deliver for the most part are highly divergent from germline
and have unusually long CDR3 regions. We now appear to have found
vector design and vector delivery conditions that allow consistent delivery
of problematic anti-SIV/anti-HIV mAbs. Nonetheless, continued work is
needed to effectively realize the full potential of this approach.
C-103
Germline-targeting vaccine design for HIV
William Schief, PhD, Scripps Research Institute
Induction of broadly neutralizing antibodies (bnAbs) against HIV is a holy
grail in the efforts to develop an HIV vaccine. The HIV envelope trimer, the
target of both bnAbs and strain-specific neutralizing antibodies, is heavily
glycosylated and has an antigenic surface that varies significantly among
different HIV isolates. Therefore, vaccine elicitation of bnAbs using stable
trimer immunogens that mimic native spikes presents major challenges.
Potent HIV bnAbs are typically highly mutated and their germline-reverted
forms typically show little or no reactivity to wild-type HIV antigens. Thus,
key barriers to bnAb elicitation include activating bnAb precursor B cells
and guiding bnAb maturation. With the ultimate aim of developing a vaccine
that consistently induces potent bnAbs in humans, we are pursuing a
strategy to develop germline-targeting priming immunogens to initiate bnAb
development, following by structure-guided boost immunogens to shepherd
B cell maturation to the development of bnAbs.
19th Annual International Meeting of the Institute of Human Virology
C-104
Defining the glycopeptide epitopes of broadly neutralizing
antibodies for HIV vaccine design
Lai-Xi Wang, PhD, University of Maryland
A major recent progress in HIV vaccine research is the discovery of a
class of glycan-reactive broadly neutralizing antibodies (bNAbs) that
target conserved N-glycans and peptide segments at the variable (V1V2
and V3) regions. Characterization of the neutralizing epitopes of these
bNAbs, represented by PG9 and the PGT series (PGT121-135), constitutes
an important step for HIV vaccine design. Mutational and structural
studies have played a key role in epitope mapping. However, the glycan
specificity and fine epitope structures of these antibodies remain to be
characterized in details. Further epitope mapping has been complicated
by the tremendous glycosylation heterogeneity of the viral envelope
glycoproteins. To address this problem, we have launched a chemical
approach that combines synthesis and antibody binding to decipher the
glycopeptide neutralizing epitopes. Using a convergent chemoenzymatic
synthetic method, we synthesized a series of V1V2 and V3 glycopeptides
corresponding to the proposed epitopes of PG9, PGT121, PGT128, and 101074. Antibody binding studies with the well-defined glycopeptides enabled
new discoveries of the glycan specificity and the requirement of the peptide
context in the antigen recognition by these bNAbs. Moreover, a synthetic
three-component V3 glycopeptide immunogen carrying a T-helper and a
TLR2 ligand was able to raise substantial glycan-dependent antibodies in
rabbits. The results will be discussed in the presentation.
C-105
Quaternary Configuration of the HIV-1 Receptor-Binding Site
Paulo Lusso, MD, PhD, National Institute of Allergy and Infectious Diseases;
Qingbo Liu, PhD, NIH; Priyamvada Acharya, PhD, NIH; Michael Dolan,
PhD, NIH; Peng Zhang, PhD, NIH; Christina Guzzo, PhD, NIH; Jacky Lu,
MS, NIH; Alice Kwon, MS, NIH; Deepali Gururani, MS, NIH; et al
The native trimeric HIV-1 envelope (Env) spike displayed on the surface
of mature virions is the sole functional form that mediates viral attachment
and entry. Interaction of conserved elements in the external envelope
glycoprotein, gp120, with the CD4 receptor is the first critical step in
the HIV-1 infectious cycle. Although the CD4-binding site in gp120 has
been extensively characterized by mutagenesis and co-crystallization
with soluble CD4, most of these studies were performed on monomeric
gp120 subunits, precluding the recognition of potential quaternary-specific
components. Here, we show that the functional HIV-1 Env trimer interacts
with CD4 through a quaternary surface formed by coalescence of the
previously defined CD4-contact region in the gp120 outer domain with a
second binding site (CD4-BS2) in the inner domain of a neighboring gp120
protomer. Disruption of CD4-BS2 in diverse HIV-1 Envs reduces the stability
of CD4-trimer interaction and abrogates HIV-1 infectivity by preventing the
acquisition of coreceptor-binding competence. A corresponding reduction
in HIV-1 infectivity occurs upon mutation of CD4 residues that interact with
CD4-BS2. A 6.8-Å cryoEM structure of a stabilized trimer (DS-SOSIP) in
complex with soluble CD4 confirmed the quaternary contact. Selected
neutralizing human antibodies also recognize a quaternary surface that
spans CD4-BS2, providing evidence that this region is immunogenic in vivo.
These results document the quaternary configuration of the functional HIV1 receptor-binding site, with implications for treatment and vaccine design.
38
Speaker Abstracts
C-106
Pan-neutralizing antibodies derived from human plasma targeting
a new epitope
Mohammad M. Sajadi,1,2 Amir Dashti,1 Marzena Pazgier,1 William D.
Tolbert,1 Michael S. Seaman,3 Xin Ouyang,1 Dongkyoon Kim,4 Guy Cavet,4
Robert R. Redfield,1 George K. Lewis,1 and Anthony L. DeVico1
Using a new methodology to obtain the sequences of antibodies in
circulation, we have discovered a family of antibodies of remarkable
breadth and potency: the N49 P series. Some members of this family have
the capability to neutralize 100% of isolates tested in a Tier 1-3 multiclade
117 pseudovirus panel, including all pseudovirsuses that were resistant
to other mAbs tested in the same panel (PGT121, GT 128, PGT 145,
PGDM1400, PGT 151, 10-107 4, 10E8, PG9, PG16, 3BNC117, NIH 45-46,
8ANC195, VRC01, and VRC07). Elisa studies and sequence analysis show
the N49 mAbs to have some characteristics of CD4-binding site antibodies.
However, crystallographic studies show a bypassing of the CD4-binding
site pocket in favor on the inner domain. The N49 P series has potential
to be useful in passive immunization and prophylactic vaccine design for
HIV-1.
1. Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
2. Department of Medicine, Baltimore VA Medical Center, Baltimore, MD 21201
3. Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard
Medical School, 3 Blackfan Circle, Boston, MA 02115
4. Atreca, Inc., Redwood City, CA 94063
D-101
The role of stress agents in immunization against HIV-1 infection
and eliciting CD4+ memory stem cells
Thomas Lehner, MD, Kings College London, University of London;
Durdana Rahman, PhD, University of London; Yufei Wang, MD, University
of London; Trevor Wittall, PhD, University of London
A robust immunological memory is critical for the longevity of any vaccine.
We have demonstrated CD4+ T stem cell memory (TSCM) in the Thai
RV144 HIV-1 vaccine clinical trial. Comparable TSCM were demonstrated
in the mucosal vaccination of women, using HIVgp140-HSP70 conjugate
administered by the vaginal route. TSCM can be upregulated by
recombinant IL-15 or DC membrane associated IL-15 (maIL-15) by a variety
of stress agents, such as thermal, oxidative or ionophore, which we studied
in vitro. Recombinant IL-15 elicited >3-fold increase and DC maIL-15 >2fold increase in CD4+ TSCM. Both systemic and mucosal immunization
elicited lower TSCM than the in vitro activated cells. Surprisingly, recall
of past exposure to immunogens, such as PPD, showed similar increase
to PPD as that to the stress agent. This was shown for CD4 proliferative
responses, Th1 cytokines and CC chemokines. As both the homeostatic
and inflammasome pathways may be stimulated by stress agents, inhibition
studies were carried out to validate this for TSCM. Indeed both pathways
may be involved in CD4+ T cell proliferation, though the homeostatic
pathway is more significant than the inflammasome one. These studies
support the concept that stress agents are important in eliciting and
maintaining CD4+ TSCM cell longevity in vaccination.
19th Annual International Meeting of the Institute of Human Virology
D-102
Immune-based interventions for HIV cure: lessons from NHP
models
Guido Silvestri, MD, Emory University
While the availability of potent anti-retroviral therapy, ART, has dramatically
reduced the mortality and morbidity associated with HIV infection, no
intervention that can functionally “cure” the infection is yet available. This is
due to a persistent reservoir of latently infected cells that is resistant to both
ART (which targets specific phases of the “productive” virus life cycle) and
immune-based interventions (which require expression of viral proteins as
target antigens). Over the past few years, the non-human primate model of
SIV/SHIV infection of rhesus macaques has been developed and validated
for studies of HIV eradication in the setting of fully suppressive ART. In this
presentation, I will review: (i) the opportunities presented by the SIV/SHIV
macaque models to conduct studies aimed at developing and testing novel
interventions to achieve a functional cure for HIV infection; (ii) the main
immune-based strategies that are currently explored to reduce or eliminate
the virus reservoir in the NHP model (i.e., shock & kill, block & lock; soothe
& schmooze; push & vanish; as well as transplant and gene therapy);
and (iii) the published and ongoing preclinical trials of immune-based
interventions that are conducted by our team in ART-treated SIV-infected
rhesus macaques with the goal of inducing a functional cure. Among the
used interventions I will discuss type I interferons, interleukin-21, FTY720,
check-point blockade inhibition (i.e., inhibitors of PD-1, CTLA4, and LAG3), CD8+ T cell depletion, CD4+ T cell depletion, and autologous stem cell
transplantation.
D-103
HIV Cure: Time to Rethink the “Shock and Kill” Strategy?
John Mellors, MD, University of Pittsburgh
The favored initial approach by many to depleting HIV reservoirs and
achieving an HIV cure was the “shock and kill” strategy, consisting of
“shocking” proviruses out of latency and killing the cells with newly
expressed viral antigens, either by direct viral-mediated cytotoxicity or
immune-mediated clearance. However, in vitro models showed that proviral
latency reversal alone did not result in death of infected cells (Shan, et al.
Immunity 2012). Ex vivo experiments revealed that only a small fraction of
proviruses (~1.5%) can be reactivated to produce virions with maximum
CD4+T-cell activation, and that <<1% are reactivated with current, small
molecule latency reversing agents (Cillo, et al. PNAS 2014). In addition,
maximum CD4+T-cell activation can cause the expansion of clones carrying
intact proviruses capable of sustained production of infectious virus (Bui,
et al. PLoS Pathogens 2017). With regard to immune-mediated killing of
infected cells, increasing antibody-dependent effector functions including
ADCC, ADCP, and ADCF is a popular approach, but a recent trial (ACTG
A5342) of two doses (40 mg/kg) of the bnMAb VRC01 revealed that is had
no effect on persistent viremia and did not decrease the number infected
cells in blood or the subset expressing viral RNA. Although many more
approaches to “shock and kill” are in development and being tested alone
and in combination, these initial data summarized above suggest that the
goal of reservoir depletion may be more difficult to achieve than anticipated
because of barriers to latency reversal and resistance of infected cells to
immune-mediated clearance.
39
Speaker Abstracts
D-104
Clonal proliferation of CD4 T cells encoding intact HIV-1.
Mathias Lichterfeld, MD, Ragon Institute
HIV-1 causes a chronic, incurable disease due to CD4 T cells that
contain replication-competent provirus but exhibit little or no active viral
gene expression, and effectively resist combination antiretroviral therapy
(cART). Such latently-infected CD4 T cells possess a remarkable longterm stability and typically persist life-long, for reasons that are not fully
understood. We have used massive single-genome, near full-length nextgeneration sequencing of HIV-1 DNA derived from unfractionated PBMC,
ex vivo-isolated CD4 T cells, and phenotypically complex memory CD4
T cells from peripheral blood and lymphoid tissues to characterize the
dynamics and underlying mechanisms supporting viral persistence. These
studies demonstrated multiple sets of independent, near full-length proviral
sequences from cART-treated individuals that were completely identical,
consistent with clonal expansion of CD4 T cells harboring intact HIV-1.
Interestingly, we found that Th1 CD4 T cells, typically responsible for antiviral
immune defense, seem to harbor the majority of such clonally-expanded
intact proviruses in cells from peripheral blood. In addition, we noted that
cells harboring clonally-expanded proviral sequences frequently expressed
cell surface markers known to protect CD4 T cells during the vulnerable
phase of clonal proliferation, suggesting that HIV-1-infected CD4 T cells
rely on physiological mechanisms for maintaining viral reservoir stability
through clonal expansion. A closer longitudinal analysis of intact proviruses
in distinct CD4 T cell subsets in future studies will be highly informative for
developing targeted interventions for viral reservoir manipulation in clinical
settings.
D-105
Reversible HIV-1 Latency Induced in Primary Human MonocyteDerived Macrophages by Repeated M1 Polarization
Francesca Graziano, PhD, Institut Curie, Paris, France; Giulia Aimola,
MS, San Raffaele Scientific Institute, Milano, Italy; Greta Forlani, PhD,
University of Insubria, Varese, Italy; Filippo Turrini, PhD, San Raffaele
University, Milano, Italy; Roberto Accolla, MD, University of Insubria,
Varese, Italy; Elisa Vicenzi, PhD, San Raffaele Scientific Institute, Milano,
Italy; Guido Poli, MD, San Raffaele University, Milano, Italy
The contribution of tissue-associated macrophages to the viral reservoir
in HIV-1 infected individuals receiving cART remains highly debated. In
this regard, we have previously reported that functional M1 polarization of
primary human MDM by short-term (18 h) stimulation with pro-inflammatory
cytokines (IFN-γ plus TNF-α) leads to a significant containment of virus
replication. Here, we demonstrate that restimulation of infected M1-MDM
with the same pro-inflammatory cytokines 7 days after infection (M12 MDM)
leads to a superior containment of virus replication to near undetectable
levels, as determined by RT activity released in culture supernatants, in
comparison to both control (CTR) MDM and to M1-MDM that were not
restimulated by polarizing cytokines. M12 MDM showed an upregulation of
APOBEC3A and 3G, with a significant reduction of HIV DNA and virtual lack
of viral mRNA expression together with the expression of transcriptional
inhibitors of proviral gene expression, namely CIITA and TRIM22,
although expression and phosphorylation of transcriptional inducers of
HIV-1 provirus, such as NF-kB and STAT-1, were not impaired. Latently
infected M12-MDM harbored replication-competent virus that was promptly
reactivated by allogeneic PHA blasts or their culture supernatant. Thus, our
study provides a formal
19th Annual International Meeting of the Institute of Human Virology
demonstration that a state of reversible latent HIV-1 infection can be
established in primary human MDM upon their repeated M1 polarization
by stimulation with pro-inflammatory cytokines before and after in vitro
infection leading to a dominance of restrictive over permissive factors.
D-106
Chromatin Functional States Correlate with the Reversal of
Latently HIV-1 Infected Primary CD4+ T cells
Emilie Battivelli, Matthew S. Dahabieh, Mohamed Abdel-Mohsen, J. Peter
Svensson, Israel Tojal Da Silva, Lilian B. Cohn, Andrea Gramatica, Steven
Deeks, Warner Greene, Satish K. Pillai, Eric Verdin
Current antiretroviral therapies (ART) do not allow for the eradication of
the human immunodeficiency virus (HIV) due to the presence of latent
proviruses in rare, long-lived resting CD4+ T cells. The main research
efforts to eliminate the viral reservoir are focused on the use of latency
reversing agents (LRAs) to force the reactivation of the latent provirus, while
maintaining ART to prevent de novo infections. Subsequently, reactivation
of HIV expression would kill reservoir cells via viral cytopathic effects and/
or immune clearance (“shock and kill” strategy). So far, no LRAs tested
in clinical trials have succeeded in reducing the reservoir. Furthermore,
there is limited understanding as to why some latent proviruses are being
induced, while others are not.
We used an improved dual-fluorescent HIV reporter (GKO), which
distinguishes productively infected cells from the latent population, to
investigate the efficacy of the “shock and kill” strategy. In addition, GKO
provides a unique opportunity to (1) explore the impact of HIV integration
site specificity on the fate of the infection, and to (2) characterize the
inducible subpopulation of latently infected cells, since it allows the isolation
of inducible latent and non-inducible latent populations from the productively
infected majority.
We first showed that our patients’ data was consistent with previously
published studies. However, we found that at most 5% of GKO latent
proviruses were reactivated. Moreover, the analysis of HIV-1 integration
sites from productively, non-inducible and inducible latently infected
populations reveals heterogeneity within the latent infections. In contrast
to non-inducible latent infections, the integration sites of inducible latent
proviruses have similar features to those of productive proviruses, thus
demonstrating a prominent role for the site of integration and its chromatin
context for the fate of the initial infection as well as for latency reversal.
Our study shows an important roadblock for the “shock and kill” approach to
reservoir eradication. Differences between inducible and permanently latent
reservoir cells suggest that complete reservoir reactivation and eradication
with LRAs may prove impossible, and that a multipronged “functional cure”
approach may be necessary.
40
Speaker Abstracts
D-107
HIV Env trimer immunogen design and modification to elicit
neutralizing antibodies
Richard Wyatt, PhD, IAVI Neutralizing Antibody Center at Scripps
A major challenge in HIV-1 vaccine design is to generate antibodies directed
toward conserved broadly neutralizing epitopes on the surface-exposed
viral envelope glycoprotein (Env). Most conserved epitopes are masked
by self N-glycans, limiting naïve B cell recognition of the underlying protein
surface following Env vaccination or during natural infection. Recently,
soluble faithful mimics of the HIV Env spike have been developed, including
the stabilized cleavage-independent NFL (native flexibly linked) trimers,
but their capacity to elicit broadly cross-reactive tier 2 (clinical isolate)
neutralizing responses has so far been limited. The conserved primary
receptor, CD4 binding site, is a known neutralizing determinant, but is
flanked by self-N-linked glycans, limiting B cell and antibody access to this
site. Here, we eliminated up to four N-glycans surrounding the CD4 binding
site of the NFL trimer without affecting trimer stability or conformation, as
demonstrated by multiple biophysical methods and EM. Using these wellordered trimers, we arrayed them at high density on synthetic liposomal
nanoparticles because, as we have shown, this multivalent trimer array
enhances B cell activation, germinal center formation and the elicitation
of HIV neutralizing antibodies. We performed immunogenicity experiments
in animal models, demonstrating that the N-glycan-deleted trimers elicited
superior neutralizing responses as a prime for fully glycosylated trimers,
compared to multiple immunizations with fully glycosylated trimers alone.
The N-glycan deleted priming also resulted in detectable cross-neutralization
of a small subset of tier 2-like viruses following boosting. The approach of
N-glycan deletion as a prime, coupled with multivalent trimer array, is a
promising means to elicit better HIV cross-neutralizing antibodies.
D-108
Innate-primed alternative signaling pathways enhance functional
mucosal mobilization of memory-like NK cells in HIV/SIV infection
Keith Reeves, PhD, Harvard Medical School
Burgeoning evidence indicates a broader functional repertoire for NK cells
beyond innate immunity including adaptive functions. Specifically, memorylike NK cells lacking the FcR γ-signaling chain (FcRΔg-NK cells) still
require antibody to grant antigen-specificity, but are pre-sensitized for rapid
mobilization against viral antigens. Interestingly, FcRΔg-NK cells require
innate priming by CMV, but execute memory-like killing against HIV and SIV
through poorly understood mechanisms.
Sixty rhCMV–, rhCMV+, and chronically SIV-infected macaques were
studied and compared to thirty naïve and untreated HIV-infected humans.
FcRΔg-NK cells were systemically distributed but, correlating with viral load,
increased four-fold in CMV+ and HIV/SIV-subjects, including in the GI tract.
Upregulated CD16 and α4β7 suggested memory-like priming is required
for both antibody-dependent functions and mucosal homing. FcRΔg-NK
cells exhibited two-fold more robust IFN-γ secretion and cytotoxicity in
the presence of antibody, but reduced expression of Helios and Eomes
— indicative of epigenetic modifications, and clustered independently
from traditional NK cells in multidimensional t-SNE. The γ-chain adaptor,
Syk was absent or dephosphorylated in FcRΔg-NK cells, but ζ-chain,
phosphorylated by adaptor Zap70, was significantly upregulated, indicating
use of the alternative ζ-chain/Zap70 pathway to achieve greater functional
potency.
19th Annual International Meeting of the Institute of Human Virology
Our work presents the first description of a combinatorial mechanism
of innate-priming and alternative signaling to explain the memorylike phenomena of NK cells mobilized against HIV/SIV. Future studies
harnessing memory-like NK cells could create exciting modalities for both
vaccine and curative therapies.
D-109
Defining a Potent New Class of Latency Reversing Agents Devoid
of Toxicity and Detrimental Cell Activation That Enhance CTL/NK
Cell Killing
Andrea Gramatica1, Roland Schwarzer1, Mauricio Montano1, Eytan Herzig1,
Thomas Packard1, and Warner C. Greene1,*
Despite long-term administration of antiretroviral therapy (ART), HIV-1
persists in a broadly distributed latent reservoir mainly comprised of resting
CD4+ T cells. Cells harboring latent provirus typically display little to no
HIV-1 gene expression and thus remain invisible to the immune system.
To achieve a durable sustained viral remission in HIV-infected patients
off ART (a functional cure), it will be important to both “reduce the size of
the reservoir and to control viral rebound by eliciting an effective immune
response capable of restraining viral spread from the smaller reservoir. This
approach will likely require the combined use of potent and safe latencyreversing agent (LRA) and a therapeutic HIV vaccine.
Our recent studies have shown that activators of the AKT/mTOR pathway
form a promising group of LRAs. Our interest has focused on two small
molecules, SB-216763 and Tideglusib that commonly inhibit glycogen
synthase kinase-3 (GSK-3). This inhibition triggers a metabolic shift to
glycolysis and results in sequential activation of mTORC2, AKT, and
mTORC1. Both compounds are known to have high tissue penetration
including the brain.
We found that these GSK-3 inhibitors potently activate latent HIV-1 in both
a tissue-based model of HIV latency formed in primary CD4 T cells, and in
CD4 T cells isolated from HIV-infected patients. In some experiments, these
agents are more potent than anti-CD3/anti-CD28 antibodies even though
the GSK-3 inhibitors do not appear to induce T cell activation (measured
by changes in CD69 and CD25 expression). Finally, in contrast to the
undesirable compromise of CTL/NK function associated with many first
generation LRAs (PKC activators, HDAC inhibitors), the GSK-3 inhibitors
actually enhance CTL and NK cell effector function.
In summary, our findings reveal GSK-3 inhibitors as an interesting new
class of potent, safe, non-cell activating LRAs. Next, it will be key to define
their activity in vivo using SIV-infected macaques on suppressive ART. The
fact that tideglusib is already in phase II human trials for myotonic dystrophy
and autism could accelerate its ultimate testing as an LRA in HIV-infected
patients.
1. Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA, USA
*Corresponding author
41
Speaker Abstracts
D-110
Synergism between CRISPR/Cas9 and LASER ART leads to
elimination of HIV-1 with no rebound in Humanized Mice
Howard Gendelman, MD1, University of Nebraska Medical Center;
Prasanta Dash, PhD1, University of Nebraska Medical Center; Benson
Edagwa, PhD1, University of Nebraska Medical Center; Santhi Gorantla,
PhD1, University of Nebraska Medical Center; Rafal Kaminski, PhD2,
Temple University; Ramona Bella, PhD student2, Temple University;
Won-Bin Young, PhD2, Temple University; Kamel Khalili, PhD2, Temple
University; et al
Treatment with long acting slow effective release (LASER ART) rilpivirine,
myristolyated dolutegravir, lamivudine and abacavir followed by CRISPRCas9 proviral DNA excision led to viral eradication in HIV-1 infected
humanized mice. Ultrasensitive nested and digital droplet PCR and RNA
scope assays failed to detect HIV-1 in blood, spleen, lung, kidney, liver,
gut-associated lymphoid tissue and brain in twenty-nine percent of infected
animals treated with the dual regimen. Excision of proviral DNA fragments
spanning the LTRs and the Gag gene were identified without identifiable off
target effects. The absence of viral rebound following cessation of ART with
no progeny virus recovery served to verify HIV-1 eradication. In contrast,
HIV-1 was readily detected in all infected animals treated with LASER ART
or CRISPR-Cas9 alone. Thus, sequential application of LASER ART and
CRISPR-Cas9 therapies administered to HIV-1 infected humanized mice
provides the proof of concept that viral sterilization is possible.
activated naive B cells, checkpoint inhibitor Tim3 on CD4 T cells and high
plasma sCD25. Positive correlations were found with frequencies of bulk
and antigen specific pTfh cells exhibiting high IL-21 and low IL-2 expression
and lacking in inflammatory cytokines (TNFa, IL17) production. Although
more deficiencies were evident in HIV+ old compared to other groups, the
differences between HIV+ and HIV− for Ab responses and immunologic
markers were maximal in young age.
E-101
Infectious Disease - Public Health Response
Boris Lushniak, MD, MPHD, University of Maryland College Park School
of Public Health
1. Department of Pharmacology and Experimental Neuroscience, University of Nebraska, Omaha,
NE
2. Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia,
PA
D-111
E-102
Influence of age on immune response to influenza vaccination in
virologically suppressed HIV infected persons
Policy, Practice and Science in Nigeria: Implementing Evidence
Based Research for HIV programming in Nigeria
Suresh Pallikkuth, PhD, University of Miami; Lesley deArmas, PhD,
University of Miami; Stefano Rinaldi, PhD, University of Miami; Varghese
Georgr, PhD, University of Miami; Rajendra Pahwa, MD, University of
Miami; Savita Pahwa, MD, University of Miami
Isaac F. Adewole, MBBS(Ib), FWACS, FMCOG, FAS, FRCOG,
DSc(Hons), Honourable Minister of Health, Federal Ministry of Health,
Abuja, Nigeria
Background: Approximately 5–20% of the US population contracts
influenza infection annually with >200,000 hospitalizations and >35,000
deaths that occur mainly in older adults. We investigated determinants of
immune response to influenza vaccine in relation to aging and concurrent
HIV infection.
Methods: 154 HIV infected (HIV+) virologically controlled persons on ART
and 161 HIV uninfected (HIV−) grouped by age as young (<40 yrs), middle
aged (40-59 yrs) and old (≥60 yrs) were given trivalent influenza vaccination.
Serological responses to influenza vaccine antigens were correlated
with cellular immune activation (IA), plasma markers of inflammation,
and immune phenotype and function of peripheral leukocytes, including
peripheral T follicular helper cells (pTffh).
Results: Absolute responders with >4 fold increase in titer to all the
vaccine antigens were fewer in HIV+ (14%) than in HIV− (32%), lower in
old compared to young, and inversely correlated with age for Ab to H1N1
and B antigen. Several immunologic markers were negative predictors
of the vaccine response in HIV infection; these included higher levels
of activated pTfh, monocytes expressing inflammatory marker CD11b,
19th Annual International Meeting of the Institute of Human Virology
Nigeria with a population of over 180 million currently has a national
estimate of 3% prevalence of HIV infection. Briefly, 3 million people are
living with HIV; 160,000 pregnant women living with HIV and pediatric HIV
infection account for about 30% of global burden. Following the assumption
of office by President Muhammadu Buhari in 2015, the country initiated
plan and began an aggressive drive in revitalizing the healthcare system
towards attainment of Universal Health Coverage in the country.
This presentation will chronicle policies, practices and evidences generated
in-country on biology of HIV infection, transmission and management as
well other non-orthodox claims of cure. The socio-cultural interpretations,
perceptions and behaviours as it affects HIV infection within Nigerian
space will be highlighted. The concluding section of the presentation will
focus on key priority areas of HIV programming and research that requires
international collaborations.
42
Speaker Abstracts
E-103
Global Health Security – Why is it important?
Kashef Ijaz MD, MPH, Principal Deputy Director, Division of Global Health
Protection, Center for Global Health, Centers for Disease Control and
Prevention, Atlanta, GA
Diseases know no boundaries; a health threat anywhere is a health threat
everywhere. New viral and bacterial pathogens continue to emerge and
in today’s tightly connected world a disease can be transported from an
isolated rural village to any major city in as little as 36 hours. The recent
Ebola epidemic clearly demonstrated that at majority of the countries
(at least 70%) are not prepared to respond to disease events. It also
emphasized the overdue and urgent need to implement core capacities
of WHO’s International Health Regulations (IHR, 2005). This would help
countries have the ability to prevent, detect and respond to infectious
disease threats at source, which will not only reduce morbidity and mortality
but also the economic impact for the developing countries as well as globally.
At present, 65 countries have committed to implement the Global Health
Security agenda that supports the implementation of IHR, 2005. Examples
from its recent accomplishments and achievements have demonstrated the
ability of countries to conduct timely disease surveillance and response to
public health events.
E-104
Virus genomes reveal factors that spread and sustained the Ebola
epidemic
F-102
Inflammation and immune modulation: tackling age-related stem
cell dysfunction
Heinrich Jasper, PhD, Buck Institute for Research on Aging
In aging and diseased tissues, regeneration and regenerative therapies
are limited by stem cell dysfunction and unfavorable tissue environments.
Promising strategies to improve success include interventions that enhance
stem cell function and that harness and boost endogenous tissue repair
mechanisms. We study stem cells and tissue repair in barrier epithelia and
the retina of Drosophila and mice to explore the causes and consequences
of age-related regenerative dysfunction. These studies have led to the
discovery of interventions targeting age-related inflammation, stem cell
proliferation, stem cell metabolism, innate immune responses, and the
commensal microbiota as strategies to enhance regeneration and extend
lifespan. I will discuss these strategies and provide perspectives for the
development of targeted interventions to improve tissue function in the
elderly. I will highlight strategies to improve stem cell activity by targeting
endogenous proliferation, differentiation and nutrient response pathways,
and strategies to improve tissue repair by modulating innate immunity
and host/commensal interactions. Combining such strategies is likely to
significantly improve tissue homeostasis and regenerative therapies in the
elderly, ultimately extending the healthy years of life.
F-103
Targeting Aberrant Transcription in Pre-Cancerous Stem Cells
Gytis Dudas, PhD, Fred Hutchinson Cancer Research Center
Ulrich G. Steidl, MD, PhD, Albert Einstein College of Medicine
The 2013–2016 West African epidemic caused by the Ebola virus was of
unprecedented magnitude, duration and impact. Here we reconstruct the
dispersal, proliferation and decline of Ebola virus throughout the region
by analysing 1,610 Ebola virus genomes, which represent over 5% of
the known cases. We test the association of geography, climate and
demography with viral movement among administrative regions, inferring
a classic ‘gravity’ model, with intense dispersal between larger and closer
populations. Despite attenuation of international dispersal after border
closures, cross-border transmission had already sown the seeds for an
international epidemic, rendering these measures ineffective at curbing the
epidemic. We address why the epidemic did not spread into neighbouring
countries, showing that these countries were susceptible to substantial
outbreaks but at lower risk of introductions. Finally, we reveal that this
large epidemic was a heterogeneous and spatially dissociated collection
of transmission clusters of varying size, duration and connectivity. These
insights will help to inform interventions in future epidemics.
Relapse continues to be the most common cause of death in acute
myeloid leukemia (AML) and many other cancers. Recent evidence has
shown that the accumulation of stepwise genetic and epigenetic changes
in tissue-specific stem cells lead to the formation of pre-cancerous/preleukemic stem cells (pre-LSC) that play a pivotal role not only in disease
origination but also in relapse. While the existence and essentiality of such
pre-cancerous cell states has been demonstrated in mice and humans,
still very little is known about the molecular mechanisms driving pre-LSC
formation and progression. We have recently performed molecular studies
of pre-leukemic cell states in mouse genetic models as well as primary
cells from patients, and discovered new transcription factors and regulatory
mechanisms in pre-LSC in myelodysplastic syndromes (MDS) and AML.
We have uncovered critical roles for several transcription factors in pre-LSC,
and found that enhancer haplodeficiency and resulting minimal reduction of
key transcription factors can be sufficient to induce pre-LSC formation and
subsequent progression to MDS and AML. Such models provide novel tools
for mechanistic study of pre-LSC and their progression to overt MDS and
AML, and for the development and testing of pharmacological approaches
to therapeutically interfere with these processes.
F-101
Regulation of Leukemogenesis by the Bone Marrow Niche
Stavroula Koustena, PhD, Columbia University
19th Annual International Meeting of the Institute of Human Virology
In summary, recent studies have started to shed light on pre-cancerous
stem cell states as the earliest origin of various malignancies including MDS
and AML, as well as molecular mechanisms driving their formation and
progression. These advances provide a basis for the specific therapeutic
targeting of pre-cancerous stem cells for the causative treatment of MDS
and AML and other cancers.
43
Speaker Abstracts
F-104
Decoding the regulatory networks of Leukemic Stem Cells through
Ubiquitylation
Chozha Rathinam, PhD, Institute of Human Virology, University of
Maryland Baltimore School of Medicine
In the recent years, it has become increasingly evident that hematopoietic
stem cells (HSCs) are directly involved in the recognition of both acute and
chronic infections. HSCs sense immune insults by both cell intrinsic, through
the pattern recognition receptors (PRRs), and extrinsic, mediated mainly by
the pro-inflammatory cytokines, mechanisms. However, prolonged exposure
of HSCs to inflammatory conditions results in defective differentiation of the
immune cells. Even though earlier studies suggested that a tight control on
inflammatory signals is essential for proper development of the immune
system, precise molecular mechanisms that control inflammatory signals in
HSCs and the effects of individual pro-inflammatory cytokines on HSC fate
decisions have not been elucidated. In this study, we utilized several animal
models including mice that either lack the ubiquitin editing enzyme-A20,
that functions as a negative regulator of NF-B, or express a constitutively
active form of IKK2, which results in augmented NF-B signals, in the
presence or absence of specific pro-inflammatory cytokines, to study the
impact of inflammation in the determination of HSC fate. Our data indicate
that deregulated NF-B signals in HSCs lead to pathologic hematopoiesis,
including a striking loss of lymphoid differentiation and skewing towards the
myeloid lineage. Furthermore, NF-B mediated lymphopenia was caused
by both cell intrinsic (changes in genetic and molecular signatures) and
extrinsic (elevated expression of pro-inflammatory cytokines) mechanisms.
At this annual meeting, we would be discussing the precise molecular
pathways through which inflammatory signals affect the development of
Normal and Leukemic Stem Cells.
G-101
Human papillomaviruses and carcinogenesis: well-established
and novel models
Massimo Tommasino, PhD, International Agency for Research on Cancer
Human papillomaviruses (HPVs) infect the cutaneous or mucosal epithelia.
More than 200 HPV types have been isolated so far, and they are classified
phylogenetically as genera and species. Persistent infections by the
mucosal high-risk (HR) HPV types from genus alpha have been clearly
associated with cancer development of the genital and upper respiratory
tracts. The products of two early genes, E6 and E7, are the major HR HPV
oncoproteins, being essential in all steps of the carcinogenic process.
They exert their functions by interacting with a large number of cellular
proteins, including the products of tumour suppressor genes, and altering
their properties. Biological and epidemiological data indicate that beta HPV
types, together with ultraviolet (UV) radiation, promote non-melanoma skin
cancer development. However, in contrast to the HR mucosal HPV types,
cutaneous beta HPV types appear to be required only at an early stage of
carcinogenesis, facilitating the accumulation of UV-induced DNA mutations.
Several findings also suggest that these HPV types and other carcinogens
may synergize in the induction of malignancies at different anatomical sites.
19th Annual International Meeting of the Institute of Human Virology
G-102
Endemic Burkitt lymphoma and infectious agents in cancer: new
results from the EMBLEM study implicating falciparum malaria
and Epstein-Barr virus in causation
Sam Mbulaiteye, MBChB, MPhil, MMed, National Cancer Institute
G-103
Regulation of HTLV-1 proviral latency
Charles Bangham, ScD, FMedSci, Imperial College London
The human leukaemia virus HTLV-1 causes chronic inflammatory disease
or an aggressive T-cell malignancy in about 10% of infected people. The
risk of these diseases is strongly correlated with the proviral load, which
frequently exceeds 10% of peripheral blood mononuclear cells. The high
proviral load is limited by a strong, chronically activated host immune
response. HTLV-1 does not release cell-free virions, but propagates both
within and between hosts by cell-to-cell contact, via the virological synapse.
Until recently, HTLV-1 was thought to be latent in vivo, and persisted chiefly
by continuous oligoclonal proliferation of about 100 clones of HTLV-1infected CD4+ T cells. However, we have shown that a typical individual
carries between 10^4 and 10^5 clones, and the proviral load – the chief
correlate of disease – is determined by the number of clones, not by
oligoclonal proliferation. We recently discovered that HTLV-1 alters host
chromatin structure in the infected cell, by binding the chromatin architectural
protein CTCF, which regulates higher-order chromatin structure and gene
expression in vertebrates. Thus, HTLV-1 does a remarkable experiment of
nature, by changing the conformation of chromatin in tens of thousands
of different ways in each infected host. Two broad questions are raised:
first, how does CTCF benefit the virus? Second, how does the change in
chromatin structure affect the host? I will show that the abnormal chromatin
looping caused by CTCF can deregulate host gene expression and so
may act as an oncogenic driver. I will also present evidence that HTLV-1
plus-strand is expressed in intense, intermittent bursts, and that expression
minus strand is not, as is currently believed, constitutive.
44
Speaker Abstracts
G-104
The Helicobacter pylori adhesin protein HopQ exploits the dimer
interface of human CEACAMs for oncoprotein translocation
Daniel A. Bonsor1, Qing Zhao2, Barbara Schmidinger2, Evelyn Weiss2,
Jingheng Wang3, Daniel Deredge4, Robert Beadenkopf1, Blaine Dow1,
Wolfgang Fischer2, Dorothy Beckett3, Patrick L. Wintrode4, Rainer Haas2,5,
Eric J. Sundberg1,6,7*
Helicobacter pylori infects half of the world’s population and strains that
encode the cag type IV secretion system for injection of the oncoprotein
CagA into host gastric epithelial cells are associated with elevated levels
of cancer. CagA translocation into host cells is dependent on interactions
between the H. pylori adhesin protein HopQ and human CEACAMs.
Here, we present high-resolution structures of numerous HopQ-CEACAM
complexes and CEACAMs in their monomeric and dimeric forms
establishing that HopQ uses a coupled folding and binding mechanism
to engage the canonical CEACAM dimerization interface for CEACAM
recognition. By combining mutagenesis with biophysical and functional
analyses, we show that the modes of CEACAM recognition by HopQ and
CEACAMs themselves are starkly different. Our data describe precise
molecular mechanisms by which microbes exploit host CEACAMs for
infection and enable future development of novel oncoprotein translocation
inhibitors and H. pylori-specific antimicrobial agents.
1-Institute of Human Virology, 6-Department of Medicine, and 7-Department of Microbiology and
Immunology, University of Maryland School of Medicine, University of Maryland, Baltimore
2-Max von Pettenkofer-Institut für Medizinische Mikrobiologie und Krankenhaushygiene, LudwigMaximilians-Universität, Pettenkoferstraße 9a, München, Germany
3-Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland
4-Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore
5-German Center for infection Research (DZIF), partner site LMU, München, Germany
*Corresponding author: esundberg@ihv.umaryland.edu
G-105
Role of Microbiome in Cancer
Giorgio Trinchiero, MD, National Cancer Institute
Commensal microorganisms colonize barrier surfaces of all multicellular
organisms, including those of humans. For more than 500 million years
commensal microorganisms and their hosts have coevolved and adapted
to each other. As a result, the commensal microbiota affects many immune
and non-immune functions of their hosts, and de facto the two together
comprise one metaorganism. The commensal microbiota communicates
with the host via biologically active molecules. Recently, it has been reported
that microbial imbalance may play a critical role in the development of
multiple diseases, such as cancer, autoimmune conditions and increased
susceptibility to infection. The commensal microbiota not only may affect
the development, progression and immune evasion of cancer but it has also
important effects on the response to cancer immune- and chemo-therapy.
Myeloid cells are a major component of the tumor microenvironment where
they play a dual role inducing anti-tumor immune responses but mostly
promoting immune evasion, tumor progression and metastases formation.
Myeloid cells respond to environmental factors including signals derived
from commensal microbes that modulate their function and reactivity thus
impacting the response to cancer therapy.
19th Annual International Meeting of the Institute of Human Virology
G-106
Carcinogenic Potential of Bacterial Biofilms
Cynthia Sears, MD, Johns Hopkins University
The colonic microbiome is hypothesized to contribute to the induction and
progression of colon cancer. While select bacterial species have been
implicated in colon carcinogenesis, recent data also suggest that bacterial
community organization and composition are carcinogenic. Studies of
paired surgical CRC samples and normal colon mucosa as well as colon
biopsies of healthy controls undergoing screening colonoscopy revealed
that sporadic colon tumors located proximal to the hepatic flexure are
characterized by invasive polymicrobial biofilms that extend to normal
colon tissue far distant from the tumor. In contrast, colon mucosal biofilms
occur much less frequently in individuals with colon cancer distal to the
hepatic flexure and in only about 15% of mucosal samples from healthy
colonoscopy controls. This talk will present an update on the intersection
of individual microbes, biofilms and mechanisms of colon carcinogenesis.
Together our studies support a model by which specific bacteria with their
virulence genes as well as microbiota organization act with host immune
responses to contribute to colon cancer pathogenesis.
G-107
HPV vaccines: where are we now, and where are we going?
Cornelia L. Trimble, MD, Johns Hopkins University School of Medicine
On a global scale, at least 20% of human malignancies are caused
by known infectious pathogens. Most are viruses; of these, human
papillomavirus (HPV) causes more cancers than any other. In populations
which do not have herd immunity because of insufficient uptake of the
preventive vaccines, HPV infections are endemic, in part, because they
are asymptomatic. In the cervix, disease is clinically indolent. In immunecompetent persons, a transition from the intraepithelial cancer precursor
lesion, cervical intraepithelial neoplasia 3 (CIN3) or high grade squamous
intraepithelial lesion (HSIL), to invasive carcinoma is thought to take on
the order of 10-15 years. Moreover, not all HSILs progress to cancer;
in a relatively short observational window, we and others have reported
spontaneous regression of biopsy-confirmed cervical HSIL in a subset of
women. The current standard of care treatment is surgical resection. In
women who are HIV seropositive, less is known. Persistent infections are
common, and the incidence of cervical cancer is three-fold that of immunecompetent women.
HPV lesions are relatively accessible, thereby providing a clinical setting in
which to better understand the immunobiology of disease, and to test proofof-principle of potential non-surgical interventions. The infectious etiology
presents an opportunity to manipulate immune responses to viral antigens.
Indeed, the preventative HPV vaccines, Gardasil and Cervarix, are nearly
(100%) effective in vaccinated persons. Immune-based therapies for
established HPV disease would address two significant clinical settings: (1)
in eradicating established cancer precursor lesions (CIN3/HSIL), and (2) in
recurrent or metastatic HPV cancers. Early studies have established proofof-principle in both. Challenges presented in eradicating HPV disease will
be discussed, as well as strategies moving forward.
45
Speaker Abstracts
G-108
Molecular Etiology of Cervical Cancer, HPV and gene methylation
Nicholas Wentzensen, MD, PhD, National Cancer Institute
Most cervical cancers are caused by persistent infections with one of a
dozen carcinogenic human papillomaviruses (HPV). The cancer risk differs
by HPV genotype, with HPV16 and HPV18 accounting for over 70% of all
cervical cancers. E6 and E7, are potent oncoproteins that inhibit apoptosis
by disabling p53 and activate the cell cycle by disrupting the pRB pathway. In
addition, the viral oncogenes cause major chromosomal instability already
at precancerous stages and may induce integration of the viral genome
into the host cells. Despite these strong oncogenic features of HPV, only
a small subset of HPV infections ever progress to precancer or cancer.
Recently, the important role of host and viral methylation in the molecular
etiology of cervical cancer has been recognized. We recently conducted
a discovery effort that yielded a set of host gene methylation markers that
showed promising performance to detect cervical precancer. Similarly,
we recently showed that regions of the HPV genome of 11 carcinogenic
types are highly methylated in women with CIN3 compared to women with
transient infections, suggesting that methylation is a general phenomenon
in the transition from infection to precancer. Methylation patterns were
found to be similar between closely related HPV genotypes. These findings
may improve our understanding of the molecular basis for viral persistence
and increase our ability to identify women who are more likely to progress
to cervical cancer. Measuring host or HPV DNA methylation could serve as
a specific marker for cervical precancer in cervical cancer screening.
H-101
Potential of Integrase-defective Lentiviral Vectors for HIV vaccine
delivery
Mary Klotman, MD, Duke University
19th Annual International Meeting of the Institute of Human Virology
H-102
The détente between herpes simplex viruses and their human
host
Bernard Roizman, ScD, University of Chicaco
The key mission of viruses is to spread within its ecological niche. To
achieve their mission viruses must multiply efficiently, be available at
appropriate body sites for transmission and yet not kill the host since
extreme virulence that kills the host curtails the spread of viruses in a
population as illustrated by SARS and Ebola viruses. HSV infects a very
large fraction of human population and is therefore a successful pathogen.
To achieve it mission the virus encodes functions of omission that fail to
block host immune responses and functions of commission that specifically
curtail its replication. The net effect is the evolution of a pathogen that
controls its replication so as to maintain a high level of contacts between
infected and uninfected individuals.
H-103
Towards a Universal Influenza Virus Vaccine
Peter Palese, PhD, Icahn School of Medicine at Mount Sinai
Despite FDA-approved vaccines and antivirals, seasonal and pandemic
influenza remains a serious threat associated with substantial morbidity and
mortality. While annual seasonal influenza virus vaccination is frequently
effective – albeit underutilized in most countries – a safe universal influenza
virus vaccine providing broad and long-lasting immunity would represent
a major breakthrough. We have developed vaccine constructs which
express chimeric hemagglutinins resulting in the redirection of the immune
response away from the immunodominant (variant) head domain of the
hemagglutinin toward the much more conserved stalk of the hemagglutinin
and the highly conserved neuraminidase. Such vaccine constructs work
well in animal challenge models and await extensive clinical trials in humans.
The mechanism by which these novel vaccines mediate protection is via
antibodies which do not rely on hemagglutination inhibitory (HI) activity but
rather on ADCC (antibody-dependent cell-mediated cytotoxicity) effects,
activation of complement and/or inhibition of virus replication through
directly binding to viral proteins. It is hoped that the universal influenza
virus vaccine based on chimeric hemagglutinins will provide long-lasting
protection against all seasonal and pandemic influenza virus strains in the
future with the possibility of eventually eliminating influenza B.
46
Speaker Abstracts
H-104
The HIV Pandemic in Sub-Saharan Africa: Challenges, Successes
and Leadership
Thomas Quinn, MD, MSc, NIAID, National Institutes of Health
There are 36.7 million people living with HIV, of whom 19.4 million (53%) live
in eastern and southern Africa. Last year, 1.8 million people became newly
infected, with 43% residing in eastern and southern Africa. South Africa has
over 7 million people infected (20% of the global total), the largest number
of infected people in any country. In 2016, 380,000 new infections occurred
in South Africa, along with 180,000 AIDS-related deaths. Currently one of
every 10 individuals are infected, and in some areas, one of every three
young women are infected. By 2005, South Africa had the highest incidence
in the world. This led to a vigorous response by the scientific community
to study and advocate for a more intensive public health and governmental
response. Drs. Quarraisha Abdool and Salim Abdool Karim responded
by building the Centre of AIDS Programme of Research in South Africa
(CAPRISA), one of the most respected research entities in the continent.
They documented very high rates of infection in young women, described
social patterns and risk factors of high transmission, identified mucosal
factors associated with increased susceptibility, and implemented the first
successful trial of Tenofovir microbicide, effective in decreasing acquisition
of both HIV and HSV-2 in women. They implemented antiretroviral access
programs responsible for a 29% decline in new infections in adults and
a 56% decline in neonatal infections since 2010. Their efforts along with
others have led to the largest number of people on antiretroviral therapy at
3.5 million people. Drs. Q. and S. Karim have demonstrated the scientific
leadership that will be necessary to fully control the HIV pandemic in South
Africa.
H-105
Reflections on the Impact of Nucleotide Antiviral Drugs
John Martin, PhD, Gilead Sciences
Antiviral nucleotide prodrugs have benefitted patients suffering from HIV
and viral hepatitis, providing for the possibility to live longer, healthier lives.
In addition, tenofovir has allowed for pre-exposure prophylaxis to prevent
HIV infection. This is an example of the pioneering work of Salim and
Quarraisha Karim.
19th Annual International Meeting of the Institute of Human Virology
H-106
Sustained ART-Free HIV Remission: Obstacles and Opportunities
Anthony Fauci, MD, NIAID/NIH
Profound and durable suppression of HIV by antiretroviral therapy (ART)
represents a major accomplishment in HIV research. However, HIV persists
in patients despite long-term ART therapy and if ART is withdrawn, the
virus almost invariably rebounds. Lifelong ART treatment is associated
with toxicity, residual chronic inflammation, and the accelerated onset of
diseases associated with aging. Therefore, alternatives to lifelong ART
are being pursued, with the goal of achieving sustained, ART-free HIV
remission. The first pathway to sustained ART-free HIV remission is to
completely eradicate the replication-competent HIV reservoir – a classic
“cure.” The second pathway is to control HIV rebound without eradication
of the virus – referred to as “sustained virologic remission.” Three potential
avenues to achieving sustained virologic remission will be discussed. The
first approach involves the preservation of natural HIV-specific natural
immunity without additional immune enhancements. A key challenge is to
determine why the time to HIV rebound following interruption of ART has
varied so widely. The second approach involves therapeutic vaccination;
in this regard, the results of a recently completed, placebo-controlled
therapeutic vaccine trial will be discussed. The third approach is the passive
transfer of HIV-specific antibodies. Recent data on the passive transfer
of broadly neutralizing anti-HIV antibodies (bNAbs) to individuals whose
viremia is suppressed by ART and the effect of these bNAbs in suppressing
viral rebound will be presented. In addition, the role of passive transfer
of anti-α4β7 antibody in inducing long-term, post-ART remissions in nonhuman primates will be discussed.
I-101
The Changing Natural History of HIV Infection: From Opportunistic
Infections to Inflammation and Chronic Diseases
Henry Masur, MD, Clinical Center, National Institutes of Health, Bethesda,
Maryland
Clinicians are well aware of the changing natural history of HIV infection in
regions where antiretroviral therapy is accessible. Clinical investigations of
HIV in the first 20 years of the epidemic focused on the diagnosis, treatment
and prevention of opportunistic infections. While there has always been
a need to improve management strategies, effective approaches were
developed for most opportunistic infections, although the quality and
duration of survival were short until antiretroviral therapy became effective
and durable. As patients have live longer, morbidity due to comorbidities
related to viral diseases (HCV, HBV, HPV, CMV), metabolic disorders,
chronic inflammation, and neoplastic processes have become more
prevalent. Health care systems have had to augment and expand their
screening programs for such morbidities, and develop more comprehensive
health care for HIV infected patients who survive with these co morbidities.
Washington, D.C. is an example of a city which is realigning its resources to
focus on these emerging comorbidities and to integrate care. Data on these
comorbidities provide insight into research challenges that must be met.
47
Speaker Abstracts
I-102
I-104
Probing mechanisms of accelerated coronary atherosclerosis and
heart disease in HIV+ people
Cardiovascular Disease in HIV and Chronic Hepatitis C-Infected
Patients
Robert G. Weiss, Allison G. Hays, Micaela Iantorno, Shashwatee
Bagchi, Gary Gerstenblith, Johns Hopkins and IHV, University of Maryland
Shashwatee Bagchi, MD, IHV, University of Maryland School of Medicine,
Division of Infectious Diseases
Since the advent of effective ART in the 1990’s, HIV+ people are living
longer and developing chronic diseases including cardiovascular disease
that is currently the cause of death in 8%-15% of HIV+ people. HIV+
people have accelerated atherosclerosis and an approximate 50%-70%
increase in the risk of myocardial infarction as compared to a comparable,
age-matched population. Several factors are thought to contribute to this
increased CAD risk including over-representation of traditional risk factors,
chronic inflammation, and vascular activation in HIV+ people. However, the
importance and interaction of these factors are very poorly understood.
Use of antiretroviral therapy has markedly reduced morbidity and
mortality associated with human immunodeficiency virus (HIV) infection.
Nonetheless, all-cause mortality rates remain high in HIV-infected patients
compared to the general population, with non-AIDS conditions comprising
almost half of deaths. Rates of coronary artery disease (CHD) are over
twice as high in HIV-infected compared to matched uninfected controls, with
rates anticipated to increase as this population ages. Factors contributing
to increased risk of CHD in HIV-infected patients remain to be clearly
elucidated. While some studies have strongly suggested that hepatitis
C (HCV) co-infection is associated with increased rates of CHD in HIVinfected patients, results in all studies have not been consistent. Since HIV,
HCV, and atherosclerosis are all associated with chronic inflammation and
immune activation, it can be challenging to understand relative disease
pathogenesis when found concurrently in individual patients. Identifying
predictors of CHD progression from overlapping pathways has the potential
to suggest novel preventive and therapeutic intervention strategies to
mitigate CHD progression. In addition, if HCV infection is confirmed to
confer additional risk for CHD among HIV-infected patients, this would
provide further justification to treat HCV early and aggressively in this
population irrespective of liver fibrosis stage.
This presentation will review the literature on this topic and discuss very
recent studies showing that HIV+ people on contemporary ART with viral
suppression have severely abnormal coronary endothelial function, a marker
of early atherosclerosis and independent predictor of future events, before
the development of heart disease. The presentation will review evidence
that inflammation may contribute to accelerated atherosclerosis in HIV+
people and discuss new studies investigating contributing mechanisms and
potential future therapies for reducing cardiovascular risk in HIV+ people.
I-103
The Metabolic Complications of HIV
Barry Peters, MBBS, MD, FRCP, Kings College London
Some conditions of metabolic origin are, at least in part, considered to
be associated or driven by HIV infection or its treatment. These include
hyperlipidaemia, lipodystrophy, type 2 diabetes, non-alcoholic fatty
liver disease (non-ALFLD), fragility fractures, and some elements of
cardiovascular disease.
The presentation will review the data on the individual contribution of HIV
and chronic hepatitis C infection on the development of cardiovascular
disease and the proposed mechanisms for these observations. Finally, new
research investigating the intersections of these three disease processes
with potential for future preventive and therapeutic interventions will be
discussed.
There are several behavioural factors, particularly diet exercise, alcohol,
smoking, and there are some antiretroviral drugs, that contribute to the high
incidence of metabolic disease in people living with HIV (PLWH). Some
fundamental biological and cellular mechanisms may be important in driving
metabolic abnormalities in PLWH. Irisin, a cleaved part of a protein encoded
by the FNDC5 (Fibronectin type III domain-containing protein 5) gene, has
been correlated in PLWH with adiposity, and inversely correlated with fat
free mass and some strength parameters. HIV may disrupt fundamental
metabolic host cell processes and further enable a preferential environment
for virion assembly. The metabolic syndrome and obesity appear to be key
for many cases of liver fibrosis in PLWH with concomitant hepatitis virus
infection. The METAFIB study found that adipokines and SCD163 (a soluble
form of Cluster of Differentiation receptor163), were significantly associated
with fibrosis of the liver.
To improve metabolic health outcomes in PLWH, we need further studies
to increase our evidence base and develop better therapies. Therapeutic
strategies include the identification of risk factors for metabolic disease
in PLWH, such as fatty liver, and screening for diabetes, affording the
opportunity to encourage behavioural change such as diet and exercise,
and the selection of appropriate antiretroviral medication.
19th Annual International Meeting of the Institute of Human Virology
48
Speaker Abstracts
I-105
Advances in HCV-Associated Hepatocellular Carcinoma
Patrizia Farci, MD, Hepatic Pathogenesis Section, Laboratory of Infectious
Diseases, NIAID
Over the past two decades, the incidence of HCC has more than tripled in
the United States, and this alarming trend is due primarily, if not exclusively,
to HCV infection. However, the role of HCV in hepatocarcinogenesis is
still unknown. Whether HCV elicits liver cancer indirectly through chronic
inflammation and fibrosis, or directly through the expression of viral proteins
in a manner analogous to other human oncogenic viruses, remains to be
established. In particular, there is limited information on the level of HCV
replication within malignant hepatocytes and the molecular interactions
between virus and tumor. We found a significant decrease in HCV RNA
in the tumor compared to surrounding non-tumorous tissues, whereas no
differences were observed in multiple areas of control non-HCC cirrhotic
livers. Diminished HCV replication was not associated with changes in
miR-122 expression. Tracking of individual variants demonstrated changes
in viral population between tumorous and non-tumorous areas, the
extent of which correlated with the decline in HCV RNA, suggesting HCV
compartmentalization within the tumor. In contrast, compartmentalization
was not observed between non-tumorous areas and serum, nor in controls
between different areas of the cirrhotic liver or between liver and serum. Our
findings indicate that HCV replication within the tumor is restricted, with viral
compartmentalization suggesting segregation of specific viral variants in
malignant hepatocytes. Our results provide new insights for understanding
the role of HCV in HCC and may give new impetus to investigate whether
malignant hepatocytes express or more likely have lost expression of
factors that restrict viral replication.
I-106
Hepatitis Viruses in HIV Infection
Kenneth Sherman, MD, PhD, University of Cincinnati College of Medicine
Viral hepatitis represents an important comorbidity among those with HIV
infection. The epidemiology and pathophysiology is often altered compared
to the general population with increased prevalence, increased risk of
chronicity, alteration of viral setpoints, and more rapid progression of hepatic
fibrosis in many co-infected individuals. Recently, outbreaks of hepatitis A
among MSM with HIV have been reported. Hepatitis B infection is often
unrecognized and represents an important contributor to morbidity through
both liver failure and increased risk of hepatocellular carcinoma. Hepatitis
C is also associated with higher rates of chronicity following acute infection,
and rapid hepatic fibrosis leading to liver related death. Treatment of HCV is
efficacious but must take into account the underlying antiretroviral regimen.
Hepatitis D may be more frequent among HBV/HIV infected persons than
has been previously recognized, and also represents an independent risk of
HCC. Hepatitis E infection can lead to chronic infections in those with HIV
and other immunosuppressed states. Standard vaccination practices that
are highly effective in the general population to prevent hepatitis A and B
are of lower utility among those with HIV and alternative vaccine strategies
should be employed.
I-107
Optimisation of a therapeutic vaccine; the importance of immune
modulation
Angus Dalgleish, MD, FMedSci, St George’s University of London; Maja
Sommerfelt, PhD, BionorPharma; Birger Sørensen, PhD, BionorPharma;
Peter Smith, PhD, St George’s University of London
Vacc-4X is a p24 (gag) peptide based vaccine, shown to reduce viral load
(HIVRNA) in HAART interrupted patients, confirmed in a randomised phase
II study, which also confirmed vaccination had no effect on the CD4 count.
A programme to improve the efficacy of Vacc4X has included peptide
modifications to increase uptake by dendritic cells and the breadth of the
responses to these peptides.
A recent study (Sørensen B et al, ARHRV 2017) showed that in 3 cohorts
of patients, antibodies to the novel heterodimeric construct designed to
enhance the immunogenicity of this region containing C5 (501-512) and
gp41 (732-44), was associated with slower disease progression and lower
viral loads, antibodies to the C5 (501-506) domain correlated with preserved
CD4 counts. It is proposed to combine this construct with the Vacc4X to
enhance efficacy.
These patient cohorts are all long term infected patients with compromised
immune responses, even on HAART. The IMiDs, including Lenalidomide,
have been shown to enhance immune responses to cell based vaccines invivo and Prevnar in clinical trials. Priming patients with Lenalidomide before
Vacc4X led to an increase in CD4 counts in a small randomised study. It is
proposed that if Lenalidomide had been given for longer, the effect would
have been more significant.
In addition, an immune modulator, M.vaccae, has been shown to reduce
the incidence of TB in a randomised study in HIV infected subjects in Africa.
M.vaccae and its commercial derivative (IMM-101) have been shown to be
ideal adjuvants for anticancer based vaccines.
It is proposed that immune modulation by Lenalidomide and IMM-101 could
enhance clinical benefit seen with Vacc4X and enhanced responses to C5.
CONCLUSION: Co-infection of hepatitis viruses with HIV leads to
alterations in natural history, as well as prevention and treatment strategies.
Care providers must be cognizant to these differences in order to provide
optimal care for the HIV-infected patient.
19th Annual International Meeting of the Institute of Human Virology
49
Speaker Abstracts
I-108
J-102
Assessing the contribution of myeloid cells to HIV-1 persistence
under ART
Decreased levels of seroreactivity in individuals subjected to
antiretroviral therapy early in acute HIV infection
Mario Stevenson, PhD, University of Miami Miller School of Medicine;
Viviane Machado, PhD, University of Miami
Mark Manak, PhD, Henry M Jackson Foundation, MHRP; Ashley Shutt,
MA, Henry M Jackson Foundation, MHRP; Leigh Eller, PhD, Henry
M Jackson Foundation, MHRP; Merlin Robb, PhD, Henry M Jackson
Foundation, MHRP; Linda Jagodzinski, PhD, Walter Reed Army Institute
for Research, MHRP; Jintanat Ananworanich, PhD, Henry M Jackson
Foundation, MHRP; Sheila Peel, PhD, Walter Reed Army Institute of
Research, MHRP
Assessing whether myeloid cells support HIV-1 persistence in the face
of effective ART represents a significant technical challenge. As a result,
there is as yet, no direct evidence that myeloid cells play any role in viral
persistence in ART-suppressed individuals. As a consequence, research on
myeloid cell reservoirs is falling into obscurity. Infection of macrophages can
only be initiated by HIV-1 variants that have the ability to use low levels of CD4
on the cell surface. Therefore, if a functional myeloid reservoir contributes
to viral persistence under effective ART, we would predict that viremia that
rebounds following analytic treatment interruption (ATI), would contain viral
variants that have a high affinity for CD4. We have developed an approach
that allows identification of low frequency macrophage-tropic variants in
rebounding viremia post ATI. Through single genome amplification (SGA),
we cloned a large number of viral envelopes from plasma of individuals who
underwent ATI. Furthermore, we have assessed whether macrophage-tropic
viruses contributed to viral rebound in individuals who exhibited prolonged
remission after receiving reduced intensity bone marrow transplant When
these envelopes were used to construct recombinant molecular clones, they
conferred the ability to fuse with, and replicate within primary macrophages.
We believe that these results provide strong evidence for the existence
of a myeloid cell reservoir in infected individuals on suppressive ART and
furthermore, that this reservoir contributes to viral rebound when ART is
interrupted. The longevity and the anatomic source of the reservoir from
which these macrophage-tropic viruses originate, is under investigation.
J-101
Current Challenges in HIV Diagnostics – Time to Revise our
Approach?
Sheila A. Peel, MSPH, PhD, US Military HIV Research Program (MHRP)
HIV infection remains a public health challenge; HIV testing serving as the
gateway to entrance into the HIV care, treatment, and prevention cascade.
Whether laboratory evidence of infection is generated by Point of Care test
algorithms or highly sophisticated 4th and 5th generation immunoassay/
supplemental serological and/or molecular confirmatory algorithms,
diagnostic accuracy, correct classification of HIV infection status, is now
challenged by the HIV field’s most notable advances in the arenas of HIV
Prevention and Intervention. Paradigms such as Test and Treat, Treatment
as Prevention, Pre-exposure Prophylaxis (PrEP), and Post-exposure
Prophylaxis (PEP) reduce viral burden, viral reservoir constitution, the
potential for onward HIV transmission, and decrease the risk of serious
AIDS related health outcomes. These highly effective strategies however,
have also been shown to cause the delay, reduction, or reversal of evolution
of serological responses to infection and/or reduce molecular markers to
below limits of detection by currently employed assay. Lack of evolution and/
or suppression of markers long used by the laboratory to generate evidence
confirming or refuting HIV infection can thus lead to misclassification of
status. This talk will focus on current HIV testing modalities, challenges
of HIV testing under the aforementioned scenarios, presentation of case
studies, and considerations for a revised approach.
19th Annual International Meeting of the Institute of Human Virology
WHO guidelines recommend antiretroviral treatment (ART) of all HIV
infected individuals regardless of CD4 count. Early treatment minimizes
the risk of HIV transmission, lowers the natural reservoir of the virus, and
reduces subsequent serious AIDS related events, but may also disrupt
emergence of HIV diagnostic markers.
High-risk individuals at early stages of HIV infection, (HIV-1 RNA positive,
Western Blot negative or indeterminate), were enrolled into an IRB
approved study to examine the outcomes of early HAART initiation. Plasma
samples collected at Week 0, 2, 12, and 24 were tested by EIA 1/2 Plus O,
HIV-1/2 Ag/Ab Combo, HIV-1/2 MultiSpot (MS), and HIV-1 Western Blot
(WB) (all from Bio-Rad, Redmond, WA). HIV Viral Load was determined
by Abbott m2000 HIV-1 RT PCR (Chicago, IL). Stage of HIV infection was
based on the Fiebig staging system.
Of individuals treated at Fiebig I and II, 63.6 and 59.1%, respectively, were
antibody negative at 12 weeks. At week 12, MS was reactive in 18.2%,
53.7% and 35.0% of individuals treated at Fiebig I, II, and Fiebig III/IV, and
decreased to 9.1%, 48.4% and 30.0%, respectively, by week 24. 52.9% of
individuals treated at Fiebig III/IV were negative at Week 24. In contrast,
all untreated individuals were highly reactive by EIA, WB, MS and RNA by
week 2-3 after infection and remained reactive thereafter.
Absence of serological markers in individuals treated early in infection
presents challenges in determining status of infection, as some individuals
under therapy may test serologically non-reactive and RNA negative, but
are infected. These findings may also have implications in monitoring
individuals on Pre-exposure Prophylactics (PrEP) by serological tests
alone.
50
Speaker Abstracts
J-103
Performance and usability of the OraQuick® HIV Self-Test, an oral
fluid based HIV self-test, in South Africa
Michael Reed, PhD, OraSure Technologies, Inc.; Mohammed Majam,
MBA, BSc, Wits Reproductive Health and HIV Institute
With 30% of persons living with HIV globally still not knowing their status,
increasing access to HIV testing using self-tests is an approach to achieving
the United Nations’ target goal of diagnosing 90% of all people living with
HIV by 2020. The OraQuick® HIV Self-Test is an oral fluid based self-test
providing results in as little as 20 minutes and is the first self-test to receive
WHO Prequalification (July 2017). WHO Prequalification allows access to
high quality, easy to use, accurate HIV self-testing in countries wishing to
broaden their HIV testing and treatment programs.
This talk describes the results and observations of studies conducted in
South Africa to demonstrate ease of use and generation of accurate results
in the hands of a lay user. The OraQuick® HIV Self-Test results were
compared to results from a 4th Gen EIA with sensitivity of 100% (n=76)
and specificity of 99.1% (n=324). A separate comprehension study was
conducted to assess lay users’ ability to understand information provided on
the outer packaging and product insert. Comprehension scores were high
99.5% (n=200) for all critical and safety related concepts. Understanding of
the next steps for positive results was 97.5% (n=200)[1].
Many studies have shown and continue to demonstrate that the OraQuick®
HIV Self-Test is a robust test that provides accurate and reliable results.
Incorporating an easy to use oral fluid test into current HIV testing strategies
can increase the number of individuals testing, especially those hard to
reach high risk populations currently not accessing testing.
J-104
Geenius HIV 1/2 Supplemental Assay: A Rapid, Reliable and
Simple System for the Confirmation and Differentiation of
Antibodies to HIV
Kathleen Shriver, PhD, Bio-Rad Laboratories; Muriel Cardona, PhD, BioRad Laboratories; Stephane Gadelle, PhD, Bio-Rad Laboratories; Patrice
Sarfati, PhD, Bio-Rad Laboratories
days vs. the benchmark. All the mixed titer and reference specimens were
correctly classified. (2) Geenius DBS correctly detected HIV-1 and HIV-2
DBS with minor exceptions. DBS seroconversions showed slightly reduced
sensitivity vs. plasma.
CONCLUSIONS: (1) The Geenius HIV-1/2 Confirmatory Assay was
accepted for the WHO list of prequalified diagnostics (March 17, 2017). (2)
Results of an investigational Geenius DBS procedure appear promising for
settings without capacity for venipuncture.
J-105
Recent developments in HIV testing in the US: Where we are,
what’s new, and where we hope to go
Kevin Delaney, MPH, PhD, US Centers for Disease Control and Prevention
In the last 30+ years, there have been significant advances in the field of
HIV testing. Improvements in test sensitivity for early or acute HIV infection
have occurred largely because test developers have targeted analytes that
appear sooner after infection. But there have also been advances in other
aspects of the tests, including things like reductions in the time to it takes for
tests to produce a result in the laboratory, and tests that can be performed
accurately outside of laboratory settings (e.g. in a park, nightclub, or a
church). While the performance of individual tests improved incrementally
over the last 30 years, from 1989 until 2014 the algorithm for laboratory
diagnosis of HIV infection remained largely unchanged. But, since 2014,
that guidance has already been updated twice as new data and new tests
continue to emerge. However, challenges have also been identified as
laboratories try to implement the recommended algorithm. In addition to
logistical issues with the algorithm, recent data have documented how PrEP
and early treatment initiation have led to new questions for HIV diagnostics.
In the face of these challenges, the landscape of available HIV tests
continues to expand and improve. Several groups are developing platforms
for single use devices that quantify HIV nucleic acid in less than an hour.
In fact, similar technologies have already been implemented for other
viral diseases. This presentation will review the latest information on HIV
test technologies, describe the challenges and opportunities that current
and emerging technologies create for HIV testing, and discuss barriers to
implementation and new opportunities for HIV prevention and care in the
context of this evolving technology.
INTRODUCTION: In 2017 Bio-Rad Laboratories completed launching
the FDA-approved Geenius HIV-1/HIV-2 Supplemental assay to replace
Multispot HIV-1/HIV-2 in the current CDC HIV Diagnostic Algorithm. The
Geenius cassette contains antibody-binding protein A, which is conjugated
to colloidal gold dye particles; HIV-1 (p31, gp160, p24, gp41) and HIV-2
(gp36, gp140) antigens are bound to the membrane solid phase. Serum,
plasma, or whole blood may be tested. Geenius uses an automated
cassette reader and proprietary software to interpret HIV-1 and HIV-2
results. We report more recent performance data for this system, as well
as an investigational protocol for testing dried blood spot (DBS) specimens.
METHODS: (1) WHO evaluated the Geenius HIV-1/HIV-2 Supplemental
testing system with a panel of 1117 specimens. (2) DBS specimens were
tested on Geenius using an investigational procedure (40 µL of DBS eluate
and one drop of assay buffer). Samples included established infections
(131 HIV-1, 31 HIV-2, one dual), 60 seroconversion specimens, and 106
DBS collected by CDC during HIV surveillance.
RESULTS: (1) Geenius sensitivity in the WHO evaluation was 100% and
specificity was 97.4%. Seroconversion samples were detected +0.875
19th Annual International Meeting of the Institute of Human Virology
51
Speaker Abstracts
J-106
Programmable nucleic acid nanoswitches for the rapid, singlestep detection of antibodies in bodily fluids
Rudy Ippodrino, PhD, Ulisse Biomed
Antibody detection plays a pivotal role in the diagnosis of pathogens and
monitoring the success of vaccine immunization. However, current serology
techniques require multiple, time-consuming washing and incubation steps,
which limit their applicability in point-of-care (POC) diagnostics and highthroughput assays. We developed here a nucleic acid nanoswitch
platform able to instantaneously measure Immunoglobulins of type G and
E (IgG and IgE) levels directly in blood serum and other bodily fluids. The
system couples the advantages of target-binding induced co-localization
and nucleic acid conformational-change nanoswitches. Due to the modular
nature of the recognition platform the method can potentially be applied
to the detection of any antibody for which an antigen can be conjugated
to a nucleic acid strand. In this work we show the sensitive, fast and costeffective detection of four different antibodies and demonstrate the possible
use of this approach for the monitoring of antibody levels in HIV+ patients
immunized with AT20 therapeutic vaccine.
19th Annual International Meeting of the Institute of Human Virology
52
Poster Abstracts
P-A1
CBF-1 promotes the establishment and maintenance of HIV
latency by recruiting Polycomb repressive complexes, PRC1 and
PRC2, at HIV LTR
Mudit Tyagi, PhD, The George Washington University; Sonia Zicari, PhD,
The George Washington University; Kalamo Farley, PhD, The George
Washington University; Lin Sun, PhD, The George Washington University;
Gary Simon, MD, PhD, The George Washington University
The type of chromatin structure around the LTR promoter of integrated
HIV provirus provides critical signals that regulate transcription during both
productive and latent HIV infections. The C-promoter binding factor-1 (CBF1) is a potent and specific inhibitor of the HIV-1 transcription, which perform
its function after binding to specific sites at HIV LTR. Here we demonstrate
that the knockdown of endogenous CBF-1 in latently infected primary CD4+ T
cells, using specific small hairpin RNAs (shRNA), resulted in the reactivation
of latent HIV proviruses. By performing Chromatin immunoprecipitation
(ChIP) assays, using latently infected primary T cells and Jurkat T-cell lines,
we demonstrated that CBF-1 induces the establishment and maintenance
of HIV latency by recruiting Polycomb Group (PcG/PRC) corepressor
complexes or Polycomb repressive complexes 1 and 2 (PRC1 and PRC2).
Knockdown of CBF-1 resulted in the dissociation of PRCs corepressor
complexes and enhanced the recruitment of RNA polymerase II (RNAP
II) at HIV LTR. Knockdown of certain components of PRC1 and PRC2
leads to the reactivation of latent proviruses. Similarly, treatment of latently
infected primary CD4+ T cells with the EZH2 inhibitor, 3-deazaneplanocin A
(DZNep), led to their reactivation.
Importance: Instead of inhibiting individual enzymes, targeting factors
such as CBF-1, which mediate the establishment of multiple repressive
epigenetic changes, could be more beneficial in designing strategies to
reactivate and subsequently eliminate latent HIV reservoirs.
P-A2
Development of anti-Tat compound: MD Simulation of the Tat/
Cyclin T1/CDK9 Complex Revealing the Hidden Catalytic Cavity
within the CDK9 Molecule Upon Tat Binding
activity as the target.
P-A3
Limitations of CD32a expression as a marker of the HIV latent
reservoir
Richard Apps, George Washington University; Nathaniel Bachtel, George
Washington University; John Huang, George Washington University;
Yanqin Ren, George Washington University; Sodsai Tovanbutra, US
Military HIV Research Program and Henry M. Jackson Foundation for the
Advancement of Military Medicine; Nittaya, Phanuphak, The Thai Red
Cross AIDS Research Center; Eugene Kroon, The Thai Red Cross AIDS
Research Center, Jintanat Ananworanich, US Military HIV Research
Program and Henry M. Jackson Foundation for the Advancement of Military
Medicine
It was recently reported that CD32a accurately marks CD4+ T cells from
peripheral blood of ART suppressed people which harbor replication
competent HIV. A direct marker of these latently infected cells may be
expected to associate with reservoir size, but we find that expression
levels of CD32 measured on CD4+ T cells isolated from the blood of 12
ART-suppressed participants did not correlate with measurements of cellassociated DNA or inducible virus in these individuals. We also examined
genetic variation in CD32a, characterizing polymorphisms in 93 virally
suppressed people on ART. Our analysis of all SNPs observed, including
those reported to impact IgG binding and HIV disease progression, found
no association of any CD32a variants with levels of HIV provirus. Given
these results we attempted to repeat the primary cell sorting experiments
described in the original report of CD32 as a marker of latently infected cells.
CD4+ T cells with highest CD32 staining were sorted by flow cytometry from
peripheral blood of ART suppressed people. These populations were not
enriched in viral DNA compared to CD4+CD32- populations, in contrast to
the original report. The lack of correlation we observe between variation
in CD32 expression or genotype and reservoir size, and failure to replicate
isolation of latent cells by sorting of CD4+ T cells based on CD32 staining,
indicate CD32 may not be a direct marker of replication competent HIV in
CD4+ T cell populations.
Takashi Okamoto, MD, PhD, Nagoya City University; Kaori Asamitsu,
PhD, Nagoya City University; Takatsugu Hirokawa, PhD, National Institute
of Advanced Industrial Science and Technology (AIST)
We applied molecular dynamics (MD) simulation to analyze the dynamic
behavior of the Tat/CycT1/CDK9 tri-molecular complex and revealed the
structural changes of P-TEFb upon Tat binding. We found that Tat could
deliberately change the local flexibility of CycT1. Although the structural
coordinates of the H1 and H2 helices did not substantially change, H1ʹ,
H2ʹ, and H3ʹ exhibited significant changes en masse. Consequently, the
CycT1 residues involved in Tat binding, namely Tat-recognition residues
(TRRs), lost their flexibility with the addition of Tat to P-TEFb. In addition,
we clarified the structural variation of CDK9 in complex with CycT1 in the
presence or absence of Tat. Interestingly, Tat addition significantly reduced
the structural variability of the T-loop, thus consolidating the structural
integrity of P-TEFb. Finally, we deciphered the formation of the hidden
catalytic cavity of CDK9 upon Tat binding. MD simulation revealed that the
PITALRE signature sequence of CDK9 flips the inactive kinase cavity of
CDK9 into the active form by connecting with Thr186, which is crucial for
its activity, thus presumably recruiting the substrate peptide such as the
C-terminal domain of RNA pol II. These findings provide vital information
for the development of effective novel anti-HIV drugs with CDK9 catalytic
19th Annual International Meeting of the Institute of Human Virology
53
Poster Abstracts
P-A4
Multimodal theranostic tests for antiretroviral drug delivery
Bhavesh Kevadiya, PhD, UNMC; Christopher Woldstad, BS, UNMC;
Brendan Ottemann, BS, UNMC; Prasanta Dash, PhD, UNMC;
Balasrinivasa Sajja, PhD, UNMC; Benjamin Lamberty, BS, UNMC;
Brenda Morsey, MS, UNMC, Ted Kocher, BS, UNMC
ABSTRACT: Long acting slow effective release antiretroviral therapy (LASER
ART) was developed to improve patient regimen adherence, prevent new
infections and facilitate drug entry into human immunodeficiency viral
reservoirs. To speed LASER ART development “multimodal imaging
theranostic nanoprobes” were created. These europium (Eu3+) doped
cobalt ferrite (CF) dolutegravir (DTG) (EuCF-DTG) nanoparticles were used
as platforms for drug-particle biodistribution. After parenteral injection of
EuCF-DTG, to rats and rhesus macaques, measured drug and cobalt levels
by florescence and magnetic resonance imaging tests were found to be
coordinate to iron levels. Moreover, EuCF-DTG was found to be a log orderof-magnitude more sensitive than equivalent amounts of particle encased
iron oxide. Folic acid decoration facilitated drug particle cell uptake. We
posit that theranostic nanoprobes can facilitate LASER ART drug delivery
and be used as part of a precision nanomedicine treatment strategy.
P-A5
HIV Infected Cells Have Depolarized Membrane Potentials and
Increased Intracellular Calcium Levels.
Robert Furler, PhD, The George Washington University
Introduction/Background:
Ion distribution between the extracellular,
cytoplasmic, and organellar spaces creates membrane potentials which
drive many of life’s processes. This bioelectric membrane potential, driven
by ion channel and pump activity, can be harnessed to allow or prevent entry
of signaling mediators like Ca2+ into the cytoplasm. Several HIV proteins
have been reported to function as ion channels or alter ion channel activity.
This activity likely influences cell fate including activation and apoptosis.
Hypothesis: HIV depolarizes the plasma membrane and alters intracellular
calcium levels. Changing the polarization of the plasma membrane would
alter the levels of HIV infection.
Methods: HIV infected cells were identified using anti-Env antibody
PG9-AF647. Membrane potential measurements were done by flow
cytometry using the DiBAC4(3) dye as previously reported. Intracellular
Ca2+ measurements were also done by flow cytometry using the Fluo-4
dye. Ionomycin and PMA were used to show the contrast in intracellular
Ca2+ levels between infected and uninfected cells. To assess the effects
of membrane potential changes on HIV replication, 200μM diazoxide was
added to cells during infections.
Results: HIV infected cells consistently had depolarized membrane
potentials in both primary cells and cell lines. Additional depolarization
increased infection. Membrane depolarization was accompanied by
increased intracellular Ca2+. Ionomycin induced a drastic difference in
Ca2+ flow between uninfected and HIV-infected cells. In uninfected cells,
ionomycin induced an influx of Ca2+ while PMA had little effect. In contrast,
both ionomycin and PMA induced a large efflux of Ca2+ from HIV infected
cells.
19th Annual International Meeting of the Institute of Human Virology
P-A6
A Possible Role for Retinoic Acid in the Functional Cure of SIV
Infections in ART/α4β7 mAb-treated Monkeys
Jianshi Yu, PhD, University of Maryland School of Pharmacy; Jace Jones,
PhD, University of Maryland School of Pharmacy; Aftab Ansari, PhD,
Emory University School of Medicine; Neil Sidell, PhD, Emory University
School of Medicine; Maureen Kane, PhD, University of Maryland School of
Pharmacy Mass Spectrometry Center
Our team of collaborators has recently reported that combining short term
antiretroviral therapy (ART) with the in vivo administration of a primatized
monoclonal antibody against the α4β7 integrin (α4β7 mAb) in SIV-infected
macaques resulted in sustained control of viremia without need for
continued therapy. We have determined that acute SIV infection caused
a rapid and sustained drop in plasma retinoic acid (RA) levels that was not
corrected upon ART-mediated control of viral replication, but was restored
soon after start of α4β7 mAb treatment. This was followed by the rebound
of certain CD8α+ NK lymphocytes, cells known to be regulated by RA.
Depletion studies of the α4β7 mAb-treated “SIV controllers” with anti-CD8α
and anti-CD8β Abs have indicated that rebound of viremia and return to
suppression tracks most closely with the depletion and reappearance of NK
and NKT cells. In parallel in vitro studies using a cell line model of latent SIV
infection (Hut78 and SIV-Hut78), we determined that SIV infection inhibited
RA production, while α4β7 mAb treatment can stimulate RA production. The
data indicated that SIV-Hut78 cells showed a marked reduction in RALDH2
expression (the key regulatory enzyme in RA biosynthesis). The retinol
chaperone protein RBP1 was also reduced in SIV-Hut78 cells. Together,
these in vivo and in vitro studies indicate that an increase in RA levels is an
early sign of the efficacy of α4β7 mAb treatment and suggests that ligation
of the α4β7 integrin leads to intra-cellular signaling events that include the
restoration of RA production by select RALDH-expressing cells, and signals
that induce NK cell activation/re-distribution that may in concert play a
fundamental role in sustained control of viremia.
54
Poster Abstracts
P-A7
HIV disease history in perinatally HIV infected adolescents with
interrupted care
Tracy Evans-Gilbert, MD, MPH, CTropMed®, Jamaica Perinatal and
Paediatric HIV/AIDS Programme, Cornwall Regional Hospital; Shelly
Ann Williams, BSc Nursing, Jamaica Perinatal and Paediatric HIV/AIDS
Programme; Gail Reid, BSc Social Work, Western Regional Health
Authority; Celia Christie, MBBS, DM Peds, MPH, FAAP, FIDSA, FRCP
(Edin), Jamaica Pediatric and perinatal HIV/AIDS Programme , University
of the West Indies
Early initiation of HAART preserves the robust immune system of perinatally
HIV infected children and limits viral reservoir size. Our aim was to study the
current immune and virological status of perinatally infected adolescents
who returned after defaulting care. A retrospective analysis was conducted
on laboratory outcomes stratified by defaulters and non-defaulters and
among age bands at HAART commencement. Among 78 patients, the
median age was 14 years [IQR 13,17]. HAART was commenced at
a median age of 6 years [IQR 3,8] with 45 (57%) at >6 years. Among
27 defaulters the median time of default was 22 months. Total time on
HAART was a mean of 8.5 years ±3 years and 56 (71%) were on protease
inhibitors. Zenith HIV RNA load was a median of 4.8 log10 copies/ml [IQR
4.2, 5.2]. Nadir CD4 cell count was 368 [IQR 61,648]. Median current
CD4 percent was 33.5% [IQR 18,46 ]. Duration of CD4 of 15-25% was 12
months [IQR 4,32] for defaulters compared with 4 months [IQR 0,8] for nondefaulters. The mean duration of viral load < 500 copies /ml was 1.48± 1.68
years among defaulters, 3.6± 3.2 years among non defaulters (p<0.0001);
3.9±3 for those commencing HAART at age 1-5 yrs, and 2.4±2.6 for >6
years (p<0.05). Seventy percent of adolescents commencing HAART at 1-5
years versus 43% at age >6 year had a current CD4 count >500 (p<0.03).
Virologic suppression was 67% in the 1-5 year band versus 27% in the >
6 year band (p <0.0001). Defaulters and non defaulters did not differ with
current virologic or immunologic markers. Late age at starting HAART had a
greater negative influence on final outcome than interrupted therapy. These
results suggest the potential in HIV remission research of interrupting
HAART after early initiation of treatment.
P-A8
Establishing tissue reservoirs for the human immunodeficiency
virus in humanized mice
Hang Su, BS, UNMC; Prasanta Dash, PhD, UNMC; Santhi Gorantla,
PhD, UNMC; Larisa Poluektova, MD, PhD, UNMC; Howard Gendelman,
MD, University of Nebraska Medical Center
but over 20% in hu-PBLs mice. IHC tests showed HIV-1p24 expressing
cells only in 14-day infected hu-HSCs mice but in all infected hu-PBLs
mice. HIV-1 RNA and DNA were observed in 14-day infected hu-HSCs
mice from spleen lung liver and gut at 106-8 copies/106 hCD45+ cells,
whereas detected in 2/5 animals (~106 copies) at early time points. They
were observed in all infected hu-PBLs mouse tissues (~106copies/106
hCD45+ cells in 3-day group and 106-8 copies in other time points). HIV-1
was detected both hu-HSCs and hu-PBLs models as early as 3 days after
infection but more robust within the latter. This may be related to various
host restriction factors. Targeting these host factors may provide stronger
clues about establishment of HIV persistence.
P-A9
Transformation of Darunavir into a long acting nanoformulated
prodrug
Mary Banoub, BS, UNMC; Aditya Bade, PhD, UNMC; JoEllyn McMillan,
PhD, UNMC; Benson Edagwa, PhD, UNMC; Howard Gendelman, MD,
UNMC; et al
Antiretroviral therapy (ART) has improved the quality and longevity of HIV-1
patients. Despite such advances limitations in drug bioavailability, resistance,
and secondary toxicities abound affecting regimen adherence. We posit that
such pharmaceutical limitations may be overcome by drug transformation
into long acting slow effective release ART (LASER ART); a pharmaceutical
approach that improves cell and tissue drug penetrance and depot
formation leading to extended dosing intervals and improved antiretroviral
responses. To this end, a hydrophobic bioreversible derivative prodrug of
darunavir (DRV) was synthesized by medicinal chemistry. Modified DRV
(MDRV) was synthesized by covalent linkage of a 14-carbon hydrophobic
fatty acid moiety to the parent drug through a hemiaminal bond. A stable
poloxamer 407 coated prodrug DRV nanoformulations (NMDRV) produced
by high-pressure homogenization. Physicochemical properties of NMDRV
and resultant particle cell uptake, antiretroviral efficacy pharmacokinetic
and biodistribution studies were performed with subsequent comparisons
made between prodrug and native drug formulations. Laboratory and
pharmacokinetic tests were performed in monocyte derived macrophages
(MDM) and BALB/c mice, respectively. NMDRV displayed up to 86 mg per
106 cells in 24 hours and retention up to 2 weeks compared to undetectable
levels in the native DRV treatment. Effective plasma DRV concentration
was detected in the prodrug arm through day 7 following a single dose of 40
mg/kg compared to undetectable levels in the parent drug treatment arm.
The results highlight opportunities for LASER ART to achieve improved
ART distribution and dissolution with limited toxicity for long-term HIV/AIDS
treatments.
Due to extraordinary challenges of early viral detection and restricted access
to anatomical sites, it remains imperative to track early temporal dynamics
of viral latency establishment. The development of humanized mice has
provided new insights into the immunology, pathogenesis, treatment,
prevention, and measured eradication. In the current study, we applied
two widely used humanized mouse models of HIV/AIDS; human CD34+
hematopoietic stem cells (HSC) and peripheral blood lymphocytes (huPBLs)-transplanted NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice, to investigate
early events in HIV latency. Twenty hu-HSC or hu-PBLs mice were injected
intraperitoneally with HIV-1ADA at 104 TCID50. Infected animals were
randomly separated into 4 groups and sacrificed at day 3, 5, 7 and 14 after
infection. Blood and tissues were harvested for immunohistochemistry and
semi-nested PCR tests for HIV-1 DNA and RNA. Peripheral CD4+ T cells
were reduced by around 10% after HIV-1 infection in hu-HSCs mice
19th Annual International Meeting of the Institute of Human Virology
55
Poster Abstracts
P-A10
P-A12
Accumulation and persistence of deleted HIV proviruses following
prolonged ART
Discordant HIV Populations with Discordant V3 Tropism in CSF
and Plasma: Implications for Establishing HIV Reservoirs
Elizabeth Anderson, B.Sci, HIV Dynamics and Replication Program,
NCI; Shawn Hill, MS, MBA, HIV Dynamics and Replication Program,
NCI; Jennifer Bell, RN, BS, CCRC, Leidos, Biomedical Research Inc;
Francesco Simonetti, MD, HIV Dynamics and Replication Program, NCI;
Catherine Rehm, BA, Lab of Immunoregulation, NIAID; Sara Jones, BS,
MBA, Leidos, Biomedical Research Inc.; Rob Gorelick, PhD, Leidos,
Biomedical Research Inc; Mary Kearney, PhD, HIV Dynamics and
Replication Program, NCI; et al
Camille Lange, MSc, PhD, National Cancer Institute (NCI), NIH, Frederick,
MD; Maura Manion, MD, National Institute of Allergy and Infectious Disease
(NIAID), NIH, Bethesda, MD; Kathy Hullsiek, PhD, Division of Biostatistics,
University of Minnesota, Minneapolis, MN; Brandon Keele, PhD, National
Cancer Institute (NCI), NIH, Frederick, MD; Helene Highbarger, PhD,
Leidos Biomedical Inc, Frederick, MD; David Meya, MD, MMed, Makerere
University, Kampala, Uganda; David Boulware, MD, MPH, CTROPMED,
Department of Medicine, University of Minnesota, Minneapolis, MN; Frank
Maldarelli, MD, PhD, National Cancer Institute (NCI), NIH, Frederick, MD;
et al
HIV-1 immunotherapy offers an alternative to cART or as a ‘functional
cure’ by eliminating reactivated viral reservoirs. Both cellular and antibody
mediated immune responses have been identified which are capable of
reducing either HIV-1 infection risk or severity of infection. We present data
on both T-cell based (Vacc4x) and antibody based (vacc-C5) vaccine antigen
under development for HIV-1 immunotherapy. Vacc4x has undergone
phase II trials demonstrating reductions in viral load and sustained antiHIV-1 T-cell responses. Additional immune modulation, such as that used
in cancer immunotherapy, may have utility in HIV-1 Immunotherapy. For
this reason we combined Vacc4x with the immune modulator Lenalidomide
both in vitro and in a phase I trial of 24 subjects in order to assess the ability
of this combination to enhance CD4+ T cell responses. The combination
was safe, well tolerated and demonstrated increases in CD4+ T-cell counts
after a two week vaccine protocol. We have identified that antibodies
specific for a heterodimeric peptide construct comprising the C5501-512
and gp41732-744 regions of the HIV envelope protein (Vacc-C5) are
correlated with slow disease progression, lower viral load and markers of
immune function in HIV-1 patients and are capable of mediating antibody
dependent cytotoxicity. We conducted a phase I/II trial of 36 ART treated
patients vaccinated with Vacc-C5 antigen. Vacc-C5 was safe, well tolerated
and increased anti-Vacc-C5 titre in a subset of patients. These data support
the continued development of Vacc4x, Lenalidomide and Vacc-C5 in an
HIV-1 immunotherapeutic setting.
19th Annual International Meeting of the Institute of Human Virology
CNS reservoirs of HIV are established early in infection and may be distinct
from HIV populations in peripheral lymphoid organs, but the mechanisms
responsible for compartmentalization are not well understood. To determine
if HIV compartmentalization occurs during CNS infections, we investigated
HIV populations in plasma and CSF in patients with active cryptococcal
meningitis (CM). HIV infected patients with CM (N=73) from the COAT,
ASTRO-CM studies, had lumbar punctures and phlebotomy. HIV RNA was
quantified in plasma and CSF prior to initiating anti-CM or antiretroviral
therapy. A subset (N=9) with CSF RNA>plasma RNA and another subset
(N=9) with plasma RNA>CSF RNA underwent single genome sequencing
(SGS) of HIV env from plasma and CSF. SGS of cellular DNA was also
performed on a subset of patients (N=5) with lymphocytic pleocytosis. 992
SG sequences were obtained. Sequences were aligned and subjected to
phylogenetic analyses, compartmentalization analyses (panmyxia) and
cell tropism of the V3 loop in env for R5/X4 predictions (GENO2PHENO).
Patients with CSF pleocytosis (WBC>5 cells/µl), had higher levels of HIV in
CSF than in plasma (p<0.05). In general, proportions of R5 and X4 variants
in CSF and plasma were similar, but there was strong discordance in 4
patients, reflecting compartmentalization. Sensitive population analyses
revealed viral compartmentalization between CSF and plasma (N=5). In
patients with pleocytosis, HIV variants in CSF-derived cells were distinct
from those in CSF, but were indistinguishable from PBMC-derived HIV.
Overall, CSF compartmentalization of HIV was detectable in the majority
of patients with CM. CSF pleocytosis does not contribute substantially to
establishing HIV populations in CNS.
56
Poster Abstracts
P-C1
Development of HIV-1 Immunotherapy with Vacc4x and Vacc-C5
Peter Smith, PhD, St Georges University of London; Maja Sommerfelt,
PhD, Bionor Pharma; Grete Stjernholm, Bsc, Bionor Pharma; Mats
Ökvist, Bsc, Bionor Pharma; Jerome Zeldis, MD, PhD, Celgene Global;
Birger Sorenson, PhD, Bionor Pharma; Dag Kvale, PhD, Department
of Infectious Diseases, Oslo University Hospital, Oslo, Norway; Angus
Dalgleish, MD, PhD, St Georges University of London; et al
HIV-1 immunotherapy offers an alternative to cART or as a ‘functional
cure’ by eliminating reactivated viral reservoirs. Both cellular and antibody
mediated immune responses have been identified which are capable of
reducing either HIV-1 infection risk or severity of infection. We present
data on both T-cell based (Vacc4x) and antibody based (vacc-C5) vaccine
antigen under development for HIV-1 immunotherapy.
Vacc4x has undergone phase II trials demonstrating reductions in viral load
and sustained anti-HIV-1 T-cell responses. Additional immune modulation,
such as that used in cancer immunotherapy, may have utility in HIV-1
Immunotherapy. For this reason we combined Vacc4x with the immune
modulator Lenalidomide both in vitro and in a phase I trial of 24 subjects
in order to assess the ability of this combination to enhance CD4+ T cell
responses. The combination was safe, well tolerated and demonstrated
increases in CD4+ T-cell counts after a two week vaccine protocol.
We have identified that antibodies specific for a heterodimeric peptide
construct comprising the C5501-512 and gp41732-744 regions of the HIV
envelope protein (Vacc-C5) are correlated with slow disease progression,
lower viral load and markers of immune function in HIV-1 patients and are
capable of mediating antibody dependent cytotoxicity. We conducted a
phase I/II trial of 36 ART treated patients vaccinated with Vacc-C5 antigen.
Vacc-C5 was safe, well tolerated and increased anti-Vacc-C5 titre in a
subset of patients.
These data support the continued development of Vacc4x, Lenalidomide
and Vacc-C5 in an HIV-1 immunotherapeutic setting.
P-C2
Protection against or delay of intrarectal SHIV acquisition by
mucosal vaccines in the absence of Env-antibody responses
Yongjun Sui, PhD, National Cancer Institute; George Lewis, PhD, Institute
of Human Virology, University of Maryland; Yichuan Wang, PhD, National
Cancer Institute; Blake Frey, BS, National Cancer Institute; Amiran
Dzutsev, PhD, National Cancer Institute; Diego Vargas-Inchaustegui,
PhD, National Cancer Institute; Venkatramanan Mohanram, PhD, National
Cancer Institute, Thomas Musich, PhD, National Cancer Institute; et al
To identify the protective mechanisms of an HIV mucosal vaccine similar
to RV144, we carried out a pilot study in which we mucosally immunized
rhesus macaques with HIV/SIV peptides, SIV-expressing MVA, and fulllength single chain protein of HIV-gp120 fused to CD4. In the macaques
that received this combination, 3/14 were sterilely protected after high-dose
SHIVSF162P4 intrarectal challenge, compared to 0/29 controls (p=0.03).
We observed mucosal antigen-specific T cell responses, but not antibody
responses. We then tested efficacy of this HIV mucosal vaccine against
repeated low-dose intrarectal challenge with SHIVSF162P4. With 21
vaccinees and 7 controls, we achieved 44% vaccine efficacy (p = 0.028).
Consistent with the pilot study, the vaccine did not induce Env-specific
antibody responses in the plasma or rectal mucosa. It induced gag- and
Env-specific T cells in colorectal mucosa/MLN, but the magnitude did not
correlate with delay of viral acquisition. Interestingly, the vaccine led to
19th Annual International Meeting of the Institute of Human Virology
accumulation of myeloid-derived suppressor cells in the PBMCs and
CD14+ monocytes in the colorectal intraepithelial compartment, which
correlated with delay of viral acquisition. The vaccine also decreased
rectal gp41 antibody, total rectal mucosal plasma cells, and gut microbiome
richness. The bacterial PCA-1 correlated with number of exposures
to achieve infection, suggesting gut microbiome’s possibly influencing
HIV susceptibility. Overall, the mucosal vaccine had different protective
mechanisms from the RV144 trial. Thus, a vaccine inducing mucosal T
cells without antibodies can protect against mucosal SHIV acquisition.
Combination with a systemically-delivered antibody-inducing vaccine might
improve protection.
P-C3
A Modified Vaccinia Ankara vector expressing Lassa virus-like
particles (MVA-LasVLP) protects mice from lethal challenge with a
Lassa-Mopeia reassortant virus.
M. Camila Guzman, BS, Institute of Human Virology, University Of Maryland
School Of Medicine; Sandra Medina-Moreno, BS, Institute of Human
Virology, University of Maryland School of Medicine; Juan Zapata, PhD,
Institute of Human Virology, University of Maryland School of Medicine; HaoTing Hsu, PhD, Institute of Human Virology; Farshad Guirakhoo, PhD,
GeoVax Labs; Arban, Domi, PhD, GeoVax Labs; Nathanael McCurley,
PhD, GeoVax Labs, Rahul Basu, PhD, GeoVax Labs; et al
The Lassa-endemic areas of West Africa, including the embattled areas
of northern Nigeria have been experiencing more frequent outbreaks of
Lassa fever (early 2016 through the summer of 2017). Consequently an
international effort has been launched to produce a Lassa vaccine, including
a test of the MVA-LASV-VLP (GEO-LM02). The MVA vector has been
developed by Dr. B. Moss’ lab at NIH in collaboration with GeoVax to deliver
a variety of vaccine antigens. The GeoVAX-MVA-HIV-VLP has been used
in human trials involving more than 500 participants, it has the capacity to
express large recombinant antigens, it elicits robust T and B cell responses,
and is relatively safe due to its inability to replicate in mammalian cells. Of
all the current Lassa vaccine candidates, GEO-LM02 is the only one that
produces VLP in vivo.
Our experiments tested the optimum route of vaccination. We used young
CBA/J mice that were challenged two weeks after vaccination with an
intracerebral (ic) dose of a Lassa-Mopeia reassortant virus. Mice were given
a single dose of GEO-LM02 by subcutaneous (sc), intraperitoneal (ip), and
intramuscular (im) routes. 100% of those given im vaccine survived the
lethal challenge, whereas the other routes were slightly less protective.
All unvaccinated mice died within 8d post challenge. A second round of
experiments confirmed the previous results and demonstrated a robust
cell-mediated immunity after a single dose im vaccination. Again 100%
of the un-vaccinated mice succumbed to lethal challenge and experienced
erosion of their blood-brain-barrier. Subsequent experiments will test the
durability of immunity in surviving mice.
57
Poster Abstracts
P-C4
Associating HIV-1 Env Trimer Structures with Functional Env
Conformational States by smFRET Analysis
Maolin Lu, PhD, Yale University; Xiaochu Ma, PhD, Yale University; Luis
Castillo-Menendez, PhD, Dana Farber Cancer Institute; Jason Gorman,
PhD, NIH; James Munro, PhD, Tufts University; Peter, Kwong, PhD, NIH;
Scott Blanchard, PhD, Weill Cornell Medical School, Joseph Sodroski,
PhD, Dana Farber Cancer Institute; Walther Mothes, PhD, Yale University,
et al
The HIV-1 envelope glycoprotein (Env) trimer mainly exists in a closed
conformation (State 1), which is driven by CD4 binding through an
intermediate conformation (State 2) to the open CD4-bound conformation
(State 3). These functional Env states can be visualized by single-molecule
Fluorescence Resonance Energy Transfer (smFRET). A breakthrough in the
structural characterization of the HIV-1 Env trimer has been the generation
of recombinant cleaved soluble gp140 SOSIP.664 trimers. Parallel
cryoelectron microscopy studies have been performed with the mature
HIV-1JR-FL Env in complex with the PGT151 neutralizing antibody. Both
approaches resulted in similar structures. It is currently generally assumed
that these structures represent the ground state of HIV-1 Env (State 1). Here
we apply smFRET to probe the conformational state of HIV-1 Env in these
constructs and antibody complexes. Fluorophores were introduced at the
identical positions in the HIV-1 Env proteins used for structural studies and
the native Env on the surface of virions, and the resulting smFRET values
compared. Surprisingly, smFRET data reveal that both the soluble gp140
SOSIP.664 and PGT151-HIV-1JR-FL Env structures correspond to the
State 2 gp120 conformation observed on the virus. Our data suggest that
the all-important structure of State 1 of HIV-1 Env, which is the target of the
majority of broadly neutralizing antibodies, remains unknown. Determining
the structure of this additional conformation observed on native virions
should allow the design of second generation immunogens that specifically
present the State 1 conformation of HIV-1 Env.
P-C5
Broadly neutralizing nanobodies selected from dromedary
immune libraries with subtype C SOSIP Env glycoproteins:
optimization and preclinical development
Sarah Kalusche, PhD Student, Georg-Speyer-Haus; Felix Lehmann,
master student, Georg-Speyer-Haus; Kathrin Koch, PhD, Georg-SpeyerHaus; Florian Klein, Professor, University of Cologne; Jonathan Torres,
PhD, The Scripps Research Institute; Robyn Stanfield, PhD, The Scripps
Research Institute; Andrew Ward, Professor, The Scripps Research
Institute; Ian Wilson, Professor, The Scripps Research Institute; Ursula
Dietrich, PhD, PI, Georg-Speyer-Haus; et al
with trimeric SOSIPs identified the CD4 binding site as the major target. A
new selection performed on a next-generation optC SOSIP.664 plus sCD4
with libraries generated at a late timepoint (7 months after the initial 7 weeks
immunization cycle) identified two new nanobodies, which are currently
being analyzed for neutralization. We further proved functionality of the
nanobodies at acidic pH (as found in the vagina) and identified nanobody
combinations resulting in increased breadth of neutralization in vitro. In
view of preventive applications at vaginal sites of HIV-1 transmission, we
are expressing the best nanobodies in a membrane-bound form and as
a secreted version from lactobacilli (L. rhamnosus), which colonize the
human vagina. Finally, nanobodies will be analyzed in a humanized mouse
model of HIV-1 infection for their HIV-neutralizing capacity in vivo.
P-D1
Differential pathogenesis of human metapneumovirus clinical
isolates in C57BL/6 mice
Yu Zhang, PhD, University of Pittsburgh; Sharon Tollefson, BS, Children’s
Hospital of Pittsburgh of UPMC; Jiuyang Xu, BS, University of Pittsburgh;
John Williams, MD, Children’s Hospital of Pittsburgh of UPMC
Introduction. Human metapneumovirus (HMPV) is a member of the
Pneumoviridae, formerly a subfamily of Paramyxoviridae. HMPV is a
leading cause of lower respiratory infection in infants, children, and adults.
The disease outcome of HMPV infection ranges from mild upper respiratory
infection to severe pneumonia. Despite the clinical and economic burden,
mechanisms of HMPV pathogenesis are not fully understood. In addition,
no licensed vaccines or anti-viral drugs are available.
Results. Most published HMPV studies use a few laboratory-adapted
strains of the A2 lineage. Here, we tested eight HMPV clinical isolates from
different genetic lineages in comparison to a laboratory reference strain in
an established C57BL/6 mouse model. The clinical isolates induced variable
disease severity. While mice infected by laboratory strain TN/94-49 (A2) did
not lose weight, mice infected by clinical strains showed significant weight
loss and greater lung histopathology. Several clinical isolates caused lethal
disease, which is unusual in mouse models of HMPV. Viral replication in
lungs was variable, but peak lung viral titer did not correlate with disease
outcome. Higher proinflammatory cytokine production in mouse lungs was
associated with more severe disease and death. Virulent clinical isolates
of HMPV exhibited diminished innate immune responses and decreased
antigen-presenting cells, suggesting active inhibition of innate immunity.
The findings were confirmed in BALB/c and DBA/2 inbred mice.
Conclusion. Our results indicate that severe disease caused by HMPV clinical
isolates was due to exuberant immune response and immunopathology.
These data suggest that distinct HMPV strains may engage host immune
mediators differently.
Nanobodies or VHH are the smallest naturally occurring antibody fragments
derived from heavy chain only antibodies from Camelidae. Due to their
physicochemical properties (high stability, high affinity and target specificity,
extended CDR3 loops and their small size allowing to enter into protein
cavities), nanobodies are very suited for preventive and therapeutic
applications. We recently selected nanobodies with broad neutralizing
capacity against primary HIV-1 strains of different subtypes from phage
immune libraries generated from dromedaries immunized with HIV-1
subtype C gp140 SOSIP Env glycoproteins (Koch et al., 2017, in press).
Two nanobodies with complementary neutralization pattern neutralized 19
out of 21 pseudoviruses in the standard TZM-bl assay. Epitope mapping
data by competition ELISAs as well as negative-stain EM reconstructions
19th Annual International Meeting of the Institute of Human Virology
58
Poster Abstracts
P-D2
TRIM22 binds to CIITA and sequesters it into nuclear bodies
containing TRIM19/PML and Cyclin T1. Implications for HIV-1
infection.
Greta Forlani, PhD, University of Insubria; Filippo Turrini, PhD, San
Raffaele Scientific Institute; Guido Poli, MD, Vita-Salute San Raffaele
University; Elisa Vicenzi, PhD, San Raffaele Scientific Institute; Roberto
Accolla, MD, PhD, University of Insubria
Host restriction factors represent a defence mechanism of the innate
immune system against viral pathogens, such as HIV-1. Many studies
focused on the interplay between restriction factors and HIV-1 have greatly
improved our knowledge about the biology of the virus and the response
of the host, but to date there is no evidence on of a possible interaction
between host restriction factors to counteract the virus. Here we show
that TRIM22 and CIITA, two cellular proteins previously shown by us to
inhibit HIV-1 proviral transcription with different mechanisms, interact in
vivo and co-localize in nuclear bodies whose formation is hierarchically
controlled by TRIM22. Importantly, TRIM19/Promyelocytic Leukemia (PML)
protein, another repressor of HIV-1 transcription also acting before proviral
integration, co-localized in these nuclear bodies upon TRIM22 expression
induced by IFN-γ. Finally, TRIM22 nuclear bodies also contained CyclinT1,
a crucial elongation factor of HIV-1 primary transcripts. These findings show
that TRIM22 nuclear bodies are a site of recruitment of factors crucial for
the regulation of HIV-1 transcription and highlight the potential existence of
a concerted action between TRIM22, CIITA and TRIM19/PML to maintain a
state of proviral latency at least in myeloid cells.
P-D4
HIV-1 Env Trimer Conformational Implications of Peptide Fusion
Inhibitor Resistance
Paul Keller, PhD, FDA; Orrianne Morrison, MS, FDA; Russell Vassell,
MS, FDA; Carol Weiss, MD, PhD, FDA
P-D6
Elevated Plasma HIV RNA level is associated with impaired
neurocognitive function among HIV-1 infected patients in Nigeria
Jibreel Jumare, MBBS, PhD, UMB; Samer El-Kamary, MD, UMB;
Laurence Magder, PhD, UMB; Laura Hungerford, PhD, UMB; Anya
Umlauf, MS, UCSD; Mariana, Cherner, PhD, UCSD; Alash’le Abimiku,
PhD, UMB, Man Charurat, PhD, UMB; et al
Introduction: Plasma HIV RNA level has been shown to correlate with HIV
disease progression, morbidity and mortality. We examined the association
between levels of plasma HIV RNA and cognitive function among patients
in Nigeria.
Methods: A total of 179 HIV-1 infected participants with available plasma HIV
RNA results and followed longitudinally for up to 2 years were included in this
study. Blood samples from participants were used for the measurement of
plasma HIV RNA and CD4+ T cell count. Utilizing demographic and practice
effect adjusted T scores obtained from a 7-domain neuropsychological test
battery, cognitive status was determined by the global deficit score (GDS)
approach, with a GDS ≥ 0.5 indicating cognitive impairment.
Results: In a longitudinal multivariable linear regression analysis, adjusting
for CD4 cell count, Beck’s depression score, age, gender, years of
education, and antiretroviral treatment status, global T scores decreased
by 0.35 per log10 increase in Plasma HIV RNA [P=.0328]. Adjusting for
the same variables in a multivariable logistic regression, the odds of
neurocognitive impairment were 30% higher per log10 increase in plasma
HIV RNA (OR: 1.28 [95% CI: 1.08, 1.51]; P=0.0048). There were statistically
significant associations for the speed of information processing, executive
and verbal fluency domains in both linear and logistic regression analyses.
Conclusion: We found a significant association between plasma HIV
RNA levels and cognitive function in both baseline (cross-sectional) and
longitudinal analyses. However, the latter was significantly attenuated, and
appeared to be driven largely by strong associations among antiretroviral
naïve individuals.
Viral entry of HIV-1 is mediated by the envelope glycoprotein (Env), which
consists of gp120 and gp41 trimer subunits. Entry begins when gp120 binds
the CD4 receptor on the host cell. This induces conformational changes that
expose the binding site for the CCR5 or CXCR4 chemokine co-receptors.
Subsequently, heptad repeat 1 (HR1) and heptad repeat 2 (HR2) of gp41
self-assemble to form a six-helix bundle (6HB) that drives membrane fusion
needed for viral entry. Previously, we identified HR1 peptide resistant
Envs with key resistance mutations in HR1 or HR2 of gp41 that impact
6HB stability. These key gp41 resistance mutations defined two resistance
pathways that were each associated with additional mutations in gp120 and
gp41. Here, we further characterized the relative contribution of individual
gp120 and gp41 mutations on Env conformational structure and sensitivity
to CD4-induced conformational changes. Mutant Envs were assessed
using a panel of conformation-dependent broadly neutralizing antibodies,
temperature sensitivity studies, and soluble CD4-mediated entry into CD4CCR5+ cells. Our data show that Envs from both resistance pathways have
relaxed (more open) trimer conformations in their native state that is primarily
mediated by individual mutations in gp120. Despite increased sensitivity to
CD4 neutralization, gp41 mutations decrease conformational reactivity to
CD4 binding by altering the transition of Envs from a fusion-competent to
the inactive form. Our findings identify gp41 residues, particularly those in
HR1, as important regulators of Env conformational transitions.
19th Annual International Meeting of the Institute of Human Virology
59
Poster Abstracts
P-D8
New insights on the human anti-HIV-1 Env antibody-mediated cell
cytotoxicity (ADCC) against HIV-1 virus: Allosteric regulation of
FcRs binding upon antigen engagement
Chiara Orlandi, PhD, Institute of Human Virology, UMB School of Medicine;
Daniel Deredge, PhD, School of Pharmacy (UMB); Krishanu Ray, PhD,
Institute of Human Virology, UMB School of Medicine; Neelakshi Gohain,
PhD, Institute of Human Virology, UMB School of Medicine; William
Tolbert, PhD, Institute of Human Virology, UMB School of Medicine;
Marzena Pazgier, PhD, Institute of Human Virology, UMB School of
Medicine; Anthony DeVico, PhD, Institute of Human Virology, UMB School
of Medicine; Patrick Wintrode, PhD, UMB School of Pharmacy; et al
HIV-1 vaccine field is rapidly evolving and a deeper knowledge of the
mechanisms of defense against HIV-1 infection is needed. In this regard,
several studies highlighted the relevance of the antibody-mediated cell
cytotoxicity (ADCC) in the context of HIV-1 infection, linking Fc-effector
functions to protection against HIV-1 acquisition. The current model
for ADCC activation is based on the concept that antibodies, bound to
their respective antigens on the surface of HIV-1 sensitized cells, form
aggregates which engage the FcRs, activating the effector cells. Our
aim is to understand the molecular basis of ADCC and to identify the
critical factors that lead to the triggering of the cytotoxicity against HIV-1
virus. Utilizing multiple approaches, such as ELISA, FCS (Fluorescence
Correlation Spectroscopy), H/DX MS (Hydrogen/Deuterium Exchange
Mass Spectroscopy) and crystallography, we studied the very first step
of ADCC activation: the monovalent binding of viral antigen. Here, we
demonstrate an allosteric regulation in anti-HIV gp120 Cluster A mAbs
resulting from immune complex (IC) formation with a monomeric gp120CD4 chimera antigen. We established that IC formation dramatically
increases the efficiency of Ab interaction to low affinity FcRs compared to
free IgG, impacting, in turn, the activation of the cytotoxicity against HIV-1
positive targets. In conclusion, we believe that monomeric antigen-antibody
IC formation might be the very first step of HIV-1-specific ADCC triggering
that likely precedes the IgG aggregation required for Fc receptors binding
and in turn, effector cell activation. This might be a mechanism that enables
the fine tuning of Fc-effector functions in vaccine regimens or HIV-1 passive
treatments.
P-E1
A novel way to test and discover new inhibitors against drug
resistant HIV-1 proteases
Ge Li, PhD, Institute of Human Virology, University of Maryland School of
Medicine; Marc Ferrer, PhD, NCATS/NIH; Richard Zhao, PhD, Institute of
Human Virology, University of Maryland School of Medicine
HIV-1 protease (PR) inhibitor (PI) is one of the most potent anti-HIV drugs.
When it is used in combination with other drugs, if could suppress HIV to
an undetectable level. However, successful treatment is often threaten by
emergence of viral drug resistance of PRs (vdrPRs).
Three vdrPRs were isolated from HIV-infected patients that carry seven
(M7PR), ten (M10PR) and eleven (M11PR) PR gene mutations, respectively.
They were expressed in a gene-inducible fission yeast system to allow the
measurement of PR-specific activities. All three vdrPRs proteolyzed natural
HIV viral substrates and conferred drug resistance to Indinavir in the fission
yeast, suggesting they maintained the same proteolytic and drug resistant
activities in the fission yeast as
19th Annual International Meeting of the Institute of Human Virology
in mammalian cells. Moreover, the viral enzymatic activities of these
vdrPRs coupled with the induction of growth inhibition and cell death, which
could potentially be used as endpoints to test the efficacy of PI activities.
In this study, five investigational PIs were used to test the utility of the PRproducing yeast system with Darunavir (DRV) as a control. All six compounds
suppressed the wildtype PR and the M7PR-mediated activities. However,
none of them suppressed activities conferred by M10PR or M11PR. The
fact that M10PR and M11PR were resistant to all of the existing PI drugs
including DRV, underscores the importance of continued searching for new
PIs against vdrPRs.
The described fission yeast cell-based system might be suitable for future
testing or discovery of new PIs through high-throughput drug screening.
Because this yeast cell-based method is function-driven. It has no
presumption of what kind of PI will be found. It has the potential to uncover
novel PIs.
P-E2
The Impact of Structured Mentor Mother Support on Retention
During the First 12 Months Postpartum among HIV Positive
Women in Rural Nigeria.
Chinenye Fan-Osuala, MPH, International Research Center of Excellence,
Institute of Human Virology Nigeria; Habib Ramadhani, MBBS, MPH, PhD,
Institute of Human Virology; Salome Erekaha, MPH, International Research
Center of Excellence, Institute of Human Virology Nigeria; Udochisom
Anaba, MPH, International Research Center of Excellence, Institute of
Human Virology Nigeria; Gift Nwanne, BSc, International Research Center
of Excellence, Institute of Human Virology Nigeria; Miriam, Bathnna, BSc,
International Research Center of Excellence, Institute of Human Virology
Nigeria; Manhattan Charurat, PhD, Institute of Human Virology, Nadia
Sam-Agudu, MD, Institute of Human Virology; et al
INTRODUCTION: With the advent of lifelong therapy, interventions that
sustain long-term engagement with PMTCT services are needed. We
evaluated the impact of a structured peer support program on postpartum
PMTCT retention among rural Nigerian women.
METHODS: This prospective cohort study enrolled HIV+ pregnant women
from 20 primary healthcare centers (PHCs). Ten PHCs with structured
mentor mother (MM) support (training, supervision, client tracking, standard
documentation & performance evaluation) were pair-matched with 10
routine unstructured peer support (PS) PHCs. Participants received viral
load at 6 months and were followed up to 12 months postpartum. Viral
suppression was defined as <20 copies/ml. Retention assessment was
based on monthly & bi-monthly clinic visits in the 1st & 2nd six month
postpartum periods respectively. Participants with ≥5 of 9 expected visits
were considered retained. A logistic regression model with generalized
estimating equation was used to evaluate the effect of PS & other factors
on retention.
RESULTS: Of 497 women enrolled, 260 & 237 were exposed to MM &
routine PS respectively. Women with MM support (aOR=6.6, 95% CI 3.4
– 13.1) & viral suppression at 6 months (aOR= 3.1, 95% CI 1.8 – 5.6)
had higher odds of retention during the 12 month postpartum period. Age,
distance from PHC, religion, gravidity, disclosure & time of diagnosis had
no effect on retention.
DISCUSSION: Structure in peer support programs improved retention.
Also, viral suppression had an independent effect on retention, indicating
a strong link between adherence & sustained engagement; both being
essential for PMTCT.
CONCLUSION: Built-in structure can significantly enhance the impact of
PS interventions on PMTCT outcomes.
60
Poster Abstracts
P-E3
Assessment of internationally-available HIV test kits for their
suitability to meet manufacturers’ claims
Niel Constantine, PhD, Institute of Human Virology; Ahmed Abouhamda,
MBBS, Institute of Human Virology; Bhavneet Athwal, BS, Institute of
Human Virology; Ahmed Saleh, PhD, University of Maryland School of
Medicine; Samantha Paulus, MS, Institute of Human Virology
Background: HIV test kits, particularly rapid HIV tests, are used throughout
the world as a primary measure to protect the blood supply and provide
diagnosis to save lives. The US government purchases large numbers of
HIV tests at considerable expense, and expects them to perform adequately.
Objective: To assess a large number and variety of HIV test kits to determine
if they meet the claims of the manufacturers.
Methods: From September 2010 through July 2017, a total of 1,123 lots of
HIV test kits from 14 manufacturers and representing 11 different tests from
26 countries, were received for evaluation at the INSTITUTE OF HUMAN
VIROLOGY. Each test kit was assessed for performance characteristics
using panels of sera (n=30 or 160) that included positives for HIV-1 and HIV2 (n=8 or 80), and negatives (n=22 or 80). Several HIV-1 positive samples
were weak positives derived from seroconversion panels (SeraCare). Test
kits were also assessed for precision using several HIV-1 weak positive
samples tested in replicate.
Results: Of the HIV test kit lots evaluated, 99.2% successfully passed the
evaluation with perfect performance. Of the 9 lots that did not pass, 3 were
found to produce high background that interfered with reading, 2 performed
inadequately with high-temperature testing, and 4 exhibited greater than
one false-positive result. In one assessment, a country had reported poor
performance of a test kit, but the test kit passed the evaluation; this resulted
in a visit to the country to assess the laboratory’s activities.
Conclusion: In our evaluation of a large number and variety of HIV test kit
lots from 14 manufacturers, nearly all performed as expected and met the
manufacturers’ claims.
P-E4
The MoMent Study: Correlates of Viral Suppression at 6 months
Postpartum among HIV-Positive Women in Rural Nigeria
Habib Ramadhani, MBBS, MPH, PhD, Institute of Human Virology;
Udochisom Anaba, MPH, Institute of Human Virology Nigeria; Christopher
Isah, HND, Institute of Human Virology Nigeria; Manhattan Charurat,
PhD, Institute of Human Virology; Nadia Sam-Agudu, MD, Institute of
Human Virology
INTRODUCTION: The risk of mother-to-child transmission of HIV (MTCT)
reduces with sustained pre & postpartum administration of suppressive
maternal ARVs. The MoMent study evaluated for maternal viral suppression
& its correlates among HIV+ Nigerian women.
METHODS: This prospective cohort study compared structured Mentor
Mother (MM) vs unstructured peer support (PS) for PMTCT outcomes.
Pregnant women were recruited at 20 matched Primary Healthcare Centers
in rural North-Central Nigeria. Structured PS included daily MM supervision,
standardized documentation, client tracking, & MM performance evaluations.
Maternal viral load (VL) was performed at 6 months postpartum; women
lost to followup were tracked back for VL tests. Viral suppression was
defined as VL <20 copies/ml. All participants were on ART for ≥6 months.
Multivariate logistic regression with generalized estimating equations was
used to account for clusters and for adjusting confounders.
RESULTS: Among 497 enrolees, 296 (59.6%) presented for VL; 273/296
19th Annual International Meeting of the Institute of Human Virology
(92.2%) had samples collected. Of the 238/273 (87.2%) with available
results, 138 (58%) were suppressed. Correlates of suppression were
structured MM support (aOR 4.9, CI 2.6-9.2); age >30 years (aOR 2.3, CI
1.0-4.9); ≥secondary education (aOR 2.0, CI 1.2-3.3); Christian religion
(aOR 1.4, CI 1.1-2.1); PI-based ART (aOR 4.6, CI 1.3-16.0), and retention
(aOR 3.7, CI 2.5-5.5). Marital/disclosure status & distance from facility were
not significant.
DISCUSSION: Organized PS, older age, education, potent ARVs &
retention supported viral suppression in our study. The role of religion is
unclear & should be further explored.
CONCLUSION: Structured PS should be targeted to young & lesser
educated women to reduce MTCT risk.
P-E5
Whole Genome Deep Sequencing of HIV Reveals Extensive
Multi-class Drug Resistance in Nigerian Patients Failing First-line
Antiretroviral Therapy
Nicaise Ndembi, MPhiL, PhD, Institute of Human Virology Nigeria; Kate
Elbouzidi, MD, Division of Infection & Immunity, University College London,
London, UK; Department of Infection & Population Health, University
College London, London, UK; Vivian Kwaghe, MD, University of Abuja
Teaching Hospital, Abuja, Nigeria; Obinna Ogbanufe, MD, US Centers for
Disease Control and Prevention, US Embassy, Abuja, Nigeria; Fati MurtalaIbrahim, MPH, Institute of Human Virology Nigeria; Man Charurat, PhD,
Institute of Human Virology, University of Maryland School of Medicine;
Patrick Dakum, MD, Institute of Human Virology Nigeria; Caroline Sabin,
MD, PhD, Department of Infection & Population Health, University College
London, London, UK; et al
BACKGROUND: Whole genome deep sequencing (WGS) could improve
understanding of treatment failure and the emergence of resistance by
revealing the distribution of mutations throughout the viral population over
time.
METHODS: Adult patients receiving 1L ART (two NRTI and one NNRTI)
at the University of Abuja Teaching Hospital, Nigeria, were included if they
had experienced virological failure (HIV-1 RNA >1000 copies/mL, at least 6
months after ART initiation, confirmed by clinician-driven testing), and had
a stored plasma sample available for WGS.
RESULTS: Sixty participants were sampled during 1L failure (73% female;
median age 30 (interquartile ratio [IQR] 28-35); median CD4+ cell count 110
cells/mm3 (IQR 63-191); median 28 months after ART initiation (IQR 1841)). At 1L failure, 57% of participants had thymidine analogue mutations
(TAMs), with 30% harbouring 3 or more TAMs, 95% had other (non-TAM)
NRTI mutations and 100% had NNRTI mutations. The most common
mutations were M184V, Y181C, G190A, K65R and K103N. Overall,
17% (61/367) of the mutations identified were low-level minority variants
(present at 2-20% of the intra-host viral population), which would not have
been detected by standard resistance testing methods, 24% (88/367) were
present at 20-90% frequency, and 59% (218/367) were dominant majority
variants representing >90% of the participant’s viral population.
CONCLUSIONS: Diverse Nigerian HIV clades exhibit multi-class drug
resistance at 1L ART failure. The predominance of high-frequency mutations
suggests that emergent resistance had become fixed in the viral population
by the time of sampling.
61
Poster Abstracts
P-E6
Hepatitis B and Hepatitis C. Viral Infections Among Pregnant
Women in Some Nigerian Major Cities: A Review
Janet Itelima, PhD, Associate Professor of Applied Microbiology University
of Jos, Nigeria
Background: Worldwide, viral hepatitis is the commonest cause of hepatitis
dysfunction in pregnancy. During pregnancy, viral hepatitis is associated
with high risk of maternal complications and has become a leading cause
of foetal death.
Aim: This review was done to assess the status of hepatitis B and hepatitis
C viral infection among pregnant women in some Nigerian major cities.
Methodology: The information used for this review was from published
works in Nigeria and elsewhere. The information was extracted over the
period of seven months from November 2015 to June 2017.
Results: In Nigeria, the prevalence of hepatitis B and Hepatitis C viral
infection is on the increase and the nation has been classified among the
group of countries endemic for the infection with about 18 million of the
populace infected. The prevalence of hepatitis B viral infection among
pregnant women in many parts of the country has been reported; with Port
Harcourt having the prevalence of (4.9%), Yenagoa (5.3%), Benin (12.5%)
Jos (10.3%, 15.9% and 23.9%), Ibadan (21.3%). Anti- HCV antibody
prevalence among pregnant women has also been reported in various parts
of Nigeria; with Benin having the prevalence of (3.6%), Yenagoa (0.5%),
Osogbo (9.2%), Enugu (14.9%), Jos (5.2%), Kaduna (11.9%), Kano (7.3%)
and Zaria (18.2%). In Nigeria, the transmission of hepatitis B and Hepatitis
C viral infections occur mainly during childhood as a result of maternalneonatal transmission and by other risk factors like blood transfusion,
sexual promiscuity, history of sharing of toothbrush, sharp objects such as
razor blades, nail cutters and scissors and instruments for pedicure and
manicure. Other modes of the viral infection common in the country include
high risk groups such as health care workers, poor socioeconomic status.
Thus, all the risk factors implicated elsewhere in the spread of the viral
infections in the general population also play role in Nigeria.
Conclusion: The prevalence of hepatitis B virus (HBV) and Hepatitis C virus
(HCV) among pregnant women in Nigeria is of intermediate endemicity.
Therefore, there is the need to institute public health measures such as
routine screening of all pregnant women’s blood and blood products for
hepatitis, personal and environmental sanitation, and the discouragement
of unsupervised injections to reduce disease burden and transmission in
the population.
P-G2
Development of engineered T-cell immmunotherapy for treating
Adult-T cell leukemia caused by HTLV-1.
Juan Zapata, Msc, PhD, Institute of Human Virology, School of Medicine,
University of Maryland Baltimore; Yutaka Tagaya, MD, PhD, Institute of
Human Virology, School of Medicine, University of Maryland Baltimore
HTLV-1 infected cells by the host immune system. In addition, anti-HTLV-1
immunity is suppressed in ATL patients. We aim to target HBZ (an antisense
protein of HTLV-1) for this purpose because; 1) the frequency of T cells
recognizing HBZ correlates well with low HTLV-1 proviral loads in contrast
to T cells against Tax-1 or Env. 2) Unlike Tax-1, HBZ is constantly expressed
by all stages of ATL cells. The challenge is that HBZ is an extremely weak
immunogen. We identified a few epitopes of HBZ that stably find to Class I
HLA molecules. We tested them using an in vivo model of ATL in humanized
mice. Immunization by HBZ showed little therapeutic effects in these mice.
DC immunization showed prolonged survival, but did not protect mice from
ATL. Thus, we grew anti-HBZ CD8 T cells by immunizing normal T cells ex
vivo, enriched them by HBZ-tetramer sorting. T-cell therapy involving antiHBZ CD8 T cells successfully protected host mice, prompting us to rely on
an “engineered T cell” strategy as novel treatment of acute/lymphomatous
ATL cases.
P-G3
Unravelling the Mechanism of Action of HLBT-100 Molecule
against Adult T-Cell Leukemia
Terry-Elinor Reid, PhD, Institute of Human Virology; Josephine Geh,
MS, Institute of Human Virology; Ngeh Toyang, PhD, Institute of Human
Virology; Juan Zapata, PhD, Institute of Human Virology; Henry Lowe,
PhD, Institute of Human Virology; Joseph, Bryant, DVM, PhD, Institute
of Human Virology; Yutaka Tagaya, MD, PhD, Institute of Human Virology
Adult T-cell leukemia-lymphoma (ATL) is a leukemia of mature CD4 T
cells that develops after long-term infection of human T-cell leukemia virus
(HTLV-1). The prognosis for patients living with the disease is poor, with
chronic ATL median survival is 2 years. Once they are diagnosed with acute/
lymphomatous ATL, patients have less than one year of life left. Current
standard of care involves chemotherapy, combination of zidovudine (AZT)
and IFN-α or Arsenic Trioxide and IFNa, antibody therapy (CD25, CCR4)
and/or allogeneic stem cell transplantation, all of which will improve the
median survival but are not effective for long term treatment. Hence there
is an unmet need to discover and develop new and effective treatments
against ATL. Strategies targeting signaling networks activated in ATL has
been tried with some success. We have been testing a novel targeting of
the JAK-STAT pathway with our novel small molecule inhibitor HLBT-100.
In vitro experiments showed that HLBT-100 enhances serine-threonine
phophorylation of STAT3 by blocking the tyrosine phosphorylation of this
molecule. In normal cells, this would allow mitochondrial translocation of
the phosphorylated STAT3 after which STAT3 sustains homeostasis of cells
through the enhancement of mitochondrial energy generation. However this
pathway seems non-functional in ATL cells. Thus, we hypothesize HLBT100 seems to preferentially induce apoptotic death of ATL cells without
damaging normal cells. We are currently validating this using a novel in vivo
mouse model of ATL.
HTLV-1 is perhaps the most oncogenic virus in humans. 5 to 10% of
individuals infected by HTLV-1 develop a fatal T-cell leukemia (ATL) after
2-3 decades of latency. Conventional chemotherapy only creates escape
mutants in ATL. Novel treatments including retroviral drugs, Arsenic Trioxide/
IFNa combination, or Allogeneic stem cell transplant have increased the
5-year survival, but there is still need to establish a cure. We chose an
immunotherapy approach because all leukemic cells express some HTLV-1
viral proteins allowing to distinguish normal and
19th Annual International Meeting of the Institute of Human Virology
62
Poster Abstracts
P-G4
Molecular characterization of hippocampal changes in an animal
model of HIV associated Primary Central Nervous System
Lymphoma
Akhil Katuri, PhD, Department of Neurology, University of Maryland;
Sanketh Andhavarapu, PhD, Department of Neurology, University of
Maryland; Girma Asemu, PhD, Animal Model Division, Institute of Human
Virology; Harry Davis, MS, Animal Model Division, Institute of Human
Virology; Joseph Bryant, DVM, MS, Animal Model Division, Institute of
Human Virology; Tapas Makar, PHD, Department of Neurology, University
of Maryland; Poorna Chander Guda, PhD, Department of Neurology,
University of Maryland
Primary Central Nervous System Lymphoma (PCNSL) is a rare form of
extra nodal non-Hodgkin’s lymphoma. HIV patients are more prone to
PCNSL. It may arise from a systemic lymphoma that seeds multiple organs,
including the brain. The predominant histology of PCNSL is large B-cell
lymphoma. Molecular studies on systemic AIDS/NHL is well characterized,
however, information on molecular studies of PCNSL is limited due in
part to accessibility of tissue and lack of animal model. To date, no animal
model has been developed, which recapitulates both histopathologic and
molecular features of this disease, including the immune phenotypic state.
We recently developed an animal model of PCNSL. The observation of a
phenotype of a HIV -1 transgenic mouse model that develops a B- cell
lymphoma that mimics the disease is seen in AIDS patients with NHL Brain
lymphoma patients. HIV associated B-cell lymphoma patients as well as
HIV infected patients suffer from cognatic impairment. Brain hippocampus
region regulates cognatic behavior. Therefore, we were interested in finding
the cellular and molecular changes in the hippocampus of these animals.
Interestingly, we identified a significant increase of infiltrating leucocytes, T
cells, B cells, and macrophages/microglial cells in the hippocampal region
of those mice. Astrogliosis is also observed. Aquaporin-4 (AQP4) is the
predominant water channel expressed by astrocytes. The regulation of AQP4
has been extensively investigated in various neuropathological conditions;
however, the functional role of AQP4 in synaptic plasticity, learning, and
memory is only beginning to be elucidated. We have explored the role of
AQP4 and its influence on hippocampus and its potential relationship with
synaptic plasticity of these mice. Recent in vitro and in vivo studies using
AQP4-null and wild-type mice, in particular, the impairment of cognatic
function observed in the hippocampus. (Szu JI and Binder DK. Front Integr
Neurosci. 2016 Feb 24; 10:8). In this context we found increased AQP4
and synaptic plasticity (by detecting synaptophysin and synapsine-1) in the
hippocampus of these animals. This suggests that AQP4 plays an important
role in regulating synaptic plasticity in the hippocampus of these mice.
Astrocytes play a role in synaptic plasticity. However, there are only a few
studies that implicate a direct relationship of AQP4 in synaptic plasticity. All
together, these studies highlight the potential influence of AQP4 in synaptic
plasticity and memory of these animals probably due to compensatory
mechanism.
19th Annual International Meeting of the Institute of Human Virology
P-G5
Differential immune response to HCV peptides as cancerprogression biomarkers of HCV-infections
AnnaLucia Tornesello1, Ulf Reimer2, Pavlo Holenya2, Tobias Knaute2,
MariaLina Tornesello1 and Franco Maria Buonaguro1
HCV infections are the main cause of chronic liver disease and in part
of lymphoproliferative disorders. Most HCV infections (>90%) determine
chronic hepatitis, 30% of which progress to liver cirrhosis and 3% annually
to Hepatocellular Carcinoma (HCC). The progression rate is mainly
articulated in low (>40 years) and high (<10 years) speed progressors, with
the latter being associated to male gender, <40 years of age, >150ml daily
alcohol consumption. Current progression markers are mainly based on
biochemical evaluation of liver damage (elevation of alanine and aspartate
transaminases) and inflammation (elevation of alpha-fetoprotein). Such
markers are not specific and elevated also for other infections (i.e. HBV
and HCMV) or metabolic disorders (i.e. steatosis). Specific HCV-related
markers would be relevant to identify HCV co-factors and to select high
priority people for direct anti-viral treatment.
To identify HCC progression markers, samples from HCV+ patients at
different infection stage have been analyzed on the HCV-peptide platform
newly developed by JPT Peptide Technologies GmbH (Germany). It covers
the complete HCV-protein arrays with >3000 overlapping 15-aminoacid-long peptides from all structural and non structural HCV proteins.
The currently available data (from 7 HCV+ asymptomatic, 5 HCV+ with
cryoglobulinemia, 9 HCV cirrhosis/HCC and 5 HCV-- patients) demonstrates
that in asymptomatic patients the level of anti-HCV is in general very low
(including anti-capsid/core proteins), while high levels of immunoresponse
anti-non-structural proteins is present in patients with liver cancer.
Confirmation of such data would support the anti-non structural response
as biomarker of cancer progression in HCV+ patients.
1. Istituto Nazionale Tumori – IRCCS Fondazione Pascale, Naples, Italy
2. JPT Peptide Technologies GmbH - Volmerstrasse 5 - 12489 Berlin, Germany
63
Poster Abstracts
P-I1
Monotherapy with Integrase Inhibitors Does Not Maintain Viral
Suppression in Humanized Mice with Chronic HIV Infection
Alonso Heredia, PhD, Institute Of Human Virology; Said Hassounah,
PhD, McGill AIDS Centre; Sandra Medina-Moreno, MS, Institute Of
Human Virology; Juan Zapata, PhD, Institute Of Human Virology; Nhut
Le, BS, Institute Of Human Virology; Yingshan, Han, PhD, McGill AIDS
Centre; James Foulke Jr, MS, Institute Of Human Virology, Charles Davis,
MD, Institute Of Human Virology; et al
Introduction and Methods: Simplification of current triple ART to dual ART
or monotherapy may suffice to maintain HIV suppression while sparing
drug toxicities. We evaluated and compared the efficacy of 20-week
monotherapy with dolutegravir or raltegravir in humanized mice (HSCNSG) infected with HIVBaL. Plasma HIV RNA was measured by qRT-PCR
(limit of detection of 150 copies/40 µL plasma) and drug levels by LC/MS/
MS. Escape viruses were genotyped and analyzed for replication capacity
and drug susceptibility in tissue culture.
Results: Drug untreated control mice maintained constant viremia throughout
the study. Virus isolates from these mice were susceptible to both raltegravir
(EC50 < 8 nM) and dolutegravir (EC50 < 1 nM). Mice treated with raltegravir
or dolutegravir had plasma drug levels comparable to those in humans.
Monotherapy with raltegravir initially suppressed HIV viremia, but failed to
maintain suppression in 4/4 mice. Viruses from raltegravir failing mice had
the G140S and Q148H/K substitutions, and were resistant to both raltegravir
(EC50 values of > 100 nM) and dolutegravir (EC50 values ranging 8.8-13.3
nM) in drug susceptibility assays using human PBMCs. Monotherapy with
dolutegravir suppressed viremia in 5/5 of mice, but viremia rebounded in
one animal after 12 weeks of treatment. The virus from this mouse had
mutations E138K, G140S, Q148H, N155H and S230R, was highly resistant
to both raltegravir (EC50 > 1000 nM) and dolutegravir (EC50 of 550 nM),
and replicated to high levels in PBMCs.
Discussion: Raltegravir or dolutegravir monotherapy does not ultimately
maintain HIV suppression in humanized mice, suggesting that dual therapy
will most likely be required for simplification of ART treatment.
11 (OR=10.93) & 7 (OR=6.53) times more likely respectively, to have HIV
negative infants compared to prophylaxis regimen.Conclusion: HAART
prior to and during pregnancy is associated with an increased likelihood of
achieving a favorable EID result.
19th Annual International Meeting of the Institute of Human Virology
64