351
|
|
352
|
Margolis DM, Garcia JV, Hazuda DJ, Haynes BF. Latency reversal and viral clearance to cure HIV-1. Science 2016; 353:aaf6517. [PMID: 27463679 DOI: 10.1126/science.aaf6517] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Research toward a cure for human immunodeficiency virus type 1 (HIV-1) infection has joined prevention and treatment efforts in the global public health agenda. A major approach to HIV eradication envisions antiretroviral suppression, paired with targeted therapies to enforce the expression of viral antigen from quiescent HIV-1 genomes, and immunotherapies to clear latent infection. These strategies are targeted to lead to viral eradication--a cure for AIDS. Paired testing of latency reversal and clearance strategies has begun, but additional obstacles to HIV eradication may emerge. Nevertheless, there is reason for optimism that advances in long-acting antiretroviral therapy and HIV prevention strategies will contribute to efforts in HIV cure research and that the implementation of these efforts will synergize to markedly blunt the effect of the HIV pandemic on society.
Collapse
Affiliation(s)
- David M Margolis
- University of North Carolina HIV Cure Center, Department of Medicine, and Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA. Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| | - J Victor Garcia
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Daria J Hazuda
- Merck Research Laboratories, White Horse Junction, PA, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Department of Medicine, and Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
353
|
CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat Immunol 2016; 17:1187-96. [DOI: 10.1038/ni.3543] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 07/29/2016] [Indexed: 12/12/2022]
|
354
|
Lifson MA, Ozen MO, Inci F, Wang S, Inan H, Baday M, Henrich TJ, Demirci U. Advances in biosensing strategies for HIV-1 detection, diagnosis, and therapeutic monitoring. Adv Drug Deliv Rev 2016; 103:90-104. [PMID: 27262924 PMCID: PMC4943868 DOI: 10.1016/j.addr.2016.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 01/01/2023]
Abstract
HIV-1 is a major global epidemic that requires sophisticated clinical management. There have been remarkable efforts to develop new strategies for detecting and treating HIV-1, as it has been challenging to translate them into resource-limited settings. Significant research efforts have been recently devoted to developing point-of-care (POC) diagnostics that can monitor HIV-1 viral load with high sensitivity by leveraging micro- and nano-scale technologies. These POC devices can be applied to monitoring of antiretroviral therapy, during mother-to-child transmission, and identification of latent HIV-1 reservoirs. In this review, we discuss current challenges in HIV-1 diagnosis and therapy in resource-limited settings and present emerging technologies that aim to address these challenges using innovative solutions.
Collapse
Affiliation(s)
- Mark A Lifson
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mehmet Ozgun Ozen
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Fatih Inci
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - ShuQi Wang
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China; Institute for Translational Medicine, Zhejiang University, Hangzhou, China
| | - Hakan Inan
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA; Medicine Faculty, Zirve University, Gaziantep, Turkey
| | - Murat Baday
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Utkan Demirci
- Demirci Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, Radiology Department, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
355
|
Ganesh A, Lemongello D, Lee E, Peterson J, McLaughlin BE, Ferre AL, Gillespie GM, Fuchs D, Deeks SG, Hunt PW, Price RW, Spudich SS, Shacklett BL. Immune Activation and HIV-Specific CD8(+) T Cells in Cerebrospinal Fluid of HIV Controllers and Noncontrollers. AIDS Res Hum Retroviruses 2016; 32:791-800. [PMID: 27019338 DOI: 10.1089/aid.2015.0313] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The central nervous system (CNS) is an important target of HIV, and cerebrospinal fluid (CSF) can provide a window into host-virus interactions within the CNS. The goal of this study was to determine whether HIV-specific CD8(+) T cells are present in CSF of HIV controllers (HC), who maintain low to undetectable plasma viremia without antiretroviral therapy (ART). CSF and blood were sampled from 11 HC, defined based on plasma viral load (VL) consistently below 2,000 copies/ml without ART. These included nine elite controllers (EC, plasma VL <40 copies/ml) and two viremic controllers (VC, VL 40-2,000 copies/ml). All controllers had CSF VL <40 copies/ml. Three comparison groups were also sampled: six HIV noncontrollers (NC, VL >10,000 copies/ml, no ART); seven individuals with viremia suppressed due to ART (Tx, VL <40 copies/ml); and nine HIV-negative controls. CD4(+) and CD8(+) T cells in CSF and blood were analyzed by flow cytometry to assess expression of CCR5, activation markers CD38 and HLA-DR, and memory/effector markers CD45RA and CCR7. HIV-specific CD8(+) T cells were quantified by major histocompatibility complex class I multimer staining. HIV-specific CD8(+) T cells were detected ex vivo at similar frequencies in CSF of HC and noncontrollers; the highest frequencies were in individuals with CD4 counts below 500 cells/μl. The majority of HIV-specific CD8(+) T cells in CSF were effector memory cells expressing CCR5. Detection of these cells in CSF suggests active surveillance of the CNS compartment by HIV-specific T cells, including in individuals with long-term control of HIV infection in the absence of therapy.
Collapse
Affiliation(s)
- Anupama Ganesh
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - Donna Lemongello
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - Evelyn Lee
- Department of Neurology, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Julia Peterson
- Department of Neurology, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Bridget E. McLaughlin
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - April L. Ferre
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - Geraldine M. Gillespie
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Dietmar Fuchs
- Division of Biological Chemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Steven G. Deeks
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Peter W. Hunt
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Richard W. Price
- Department of Neurology, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Serena S. Spudich
- Department of Neurology, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| |
Collapse
|
356
|
Deleage C, Turkbey B, Estes JD. Imaging lymphoid tissues in nonhuman primates to understand SIV pathogenesis and persistence. Curr Opin Virol 2016; 19:77-84. [PMID: 27490446 PMCID: PMC5021606 DOI: 10.1016/j.coviro.2016.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/01/2016] [Accepted: 07/03/2016] [Indexed: 02/04/2023]
Abstract
CD4+ T cells are the primary HIV-1 target cell, with the vast majority of these cells residing within lymphoid tissue compartments throughout the body. Predictably, HIV-1 infection, replication, localization, reservoir establishment and persistence, as well as associated host immune and inflammatory responses and disease pathology principally take place within the tissues of the immune system. By virture of the fact that the virus-host struggle is played out within lymphoid and additional tissues compartments in HIV-1 infected individuals it is critical to understand HIV-1 infection and disease within these relevant tissue sites; however, there are obvious limitations to studying these dynamic processes in humans. Nonhuman primate (NHP) research has provided a vital bridge between basic and preclinical research and clinical studies, with experimental SIV infection of NHP models offering unique opportunities to understand key processes of HIV-1 infection and disease that are either not practically feasible or ethical in HIV-1 infected humans. In this review we will discuss current approaches to studying the tissue based immunopathogenesis of AIDS virus infection in NHPs, including both analyses of tissues obtained at biopsy or necropsy and complementary non-invasive imaging approaches that may have practical utility in monitoring HIV-1 disease in the clinical setting.
Collapse
Affiliation(s)
- Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, MD 21702, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, MD 20814, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, MD 21702, USA.
| |
Collapse
|
357
|
Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P, Chomont N, Douek D, Lifson JD, Lo YR, Kuritzkes D, Margolis D, Mellors J, Persaud D, Tucker JD, Barre-Sinoussi F, Alter G, Auerbach J, Autran B, Barouch DH, Behrens G, Cavazzana M, Chen Z, Cohen ÉA, Corbelli GM, Eholié S, Eyal N, Fidler S, Garcia L, Grossman C, Henderson G, Henrich TJ, Jefferys R, Kiem HP, McCune J, Moodley K, Newman PA, Nijhuis M, Nsubuga MS, Ott M, Palmer S, Richman D, Saez-Cirion A, Sharp M, Siliciano J, Silvestri G, Singh J, Spire B, Taylor J, Tolstrup M, Valente S, van Lunzen J, Walensky R, Wilson I, Zack J. International AIDS Society global scientific strategy: towards an HIV cure 2016. Nat Med 2016; 22:839-50. [PMID: 27400264 PMCID: PMC5322797 DOI: 10.1038/nm.4108] [Citation(s) in RCA: 364] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023]
Abstract
Antiretroviral therapy is not curative. Given the challenges in providing lifelong therapy to a global population of more than 35 million people living with HIV, there is intense interest in developing a cure for HIV infection. The International AIDS Society convened a group of international experts to develop a scientific strategy for research towards an HIV cure. This Perspective summarizes the group's strategy.
Collapse
Affiliation(s)
- Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Anna Laura Ross
- International and Scientific Relations Office, ANRS, Paris, France
| | - Jintanat Ananworanich
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Monsef Benkirane
- Molecular Virology Lab, Institute of Human Genetics, CNRS UPR 1142, Université de Montpellier, Montpellier, France
| | - Paula Cannon
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nicolas Chomont
- CRCHUM and Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, Montréal, Quebec, Canada
| | - Daniel Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Ying-Ru Lo
- World Health Organization Regional Office for the Western Pacific, Manila, Philippines
| | | | - David Margolis
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - John Mellors
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Persaud
- Johns Hopkins University School of Medicine &Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Joseph D Tucker
- University of North Carolina-Project China, Guangzhou, China
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Judith Auerbach
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Brigitte Autran
- Sorbonne Universités, UPMC Univ Paris 06, CIMI-Paris, France
- Inserm U1135, CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Georg Behrens
- Clinic for Immunology and Rhematology, Hannover Medical School, Hanover, Germany
| | - Marina Cavazzana
- Centre d'Investigation Clinique en biothérapie, Hôpital Necker-Enfants Malades, Paris, France
| | - Zhiwei Chen
- AIDS Institute, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Éric A Cohen
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Quebec, Canada
| | | | - Serge Eholié
- Programme PAC-CI, Centre Hospitalier Universitaire de Treichville, Abidjan, Côte d'Ivoire
| | - Nir Eyal
- Harvard T. H. Chan School of Public Health, Department of Global Health and Population, Boston, Massachusetts, USA
| | - Sarah Fidler
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Cynthia Grossman
- National Institute of Mental Health, NIH, Bethesda, Maryland, USA
| | - Gail Henderson
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Timothy J Henrich
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Brigham &Women's Hospital, Boston, Massachusetts, USA
| | | | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joseph McCune
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Keymanthri Moodley
- Centre for Medical Ethics and Law, Department of Medicine, Stellenbosch University, Western Cape, South Africa
| | - Peter A Newman
- Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, Ontario, Canada
| | - Monique Nijhuis
- Department of Medical Microbiology, Virology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Melanie Ott
- Gladstone Institutes, University of California, San Francisco, San Francisco, California, USA
| | - Sarah Palmer
- Westmead Millennium Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Douglas Richman
- Virginia San Diego Healthcare System and University of California, San Diego, San Diego, California, USA
| | | | - Matthew Sharp
- Independent HIV Education and Advocacy Consultant, San Francisco, California, USA
| | - Janet Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guido Silvestri
- Yerkes National Primate Research Centre, Emory University, Atlanta, Georgia, USA
| | - Jerome Singh
- Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Jeffrey Taylor
- CARE Collaboratory Community Advisory Board, Palm Springs, California, USA
| | - Martin Tolstrup
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Susana Valente
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, Jupiter, Florida, USA
| | | | - Rochelle Walensky
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ira Wilson
- Department of Health Services, Policy &Practice, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Jerome Zack
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
358
|
Notwithstanding Circumstantial Alibis, Cytotoxic T Cells Can Be Major Killers of HIV-1-Infected Cells. J Virol 2016; 90:7066-7083. [PMID: 27226367 PMCID: PMC4984658 DOI: 10.1128/jvi.00306-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Several experiments suggest that in the chronic phase of human immunodeficiency virus type 1 (HIV-1) infection, CD8+ cytotoxic T lymphocytes (CTL) contribute very little to the death of productively infected cells. First, the expected life span of productively infected cells is fairly long, i.e., about 1 day. Second, this life span is hardly affected by the depletion of CD8+ T cells. Third, the rate at which mutants escaping a CTL response take over the viral population tends to be slow. Our main result is that all these observations are perfectly compatible with killing rates that are much faster than one per day once we invoke the fact that infected cells proceed through an eclipse phase of about 1 day before they start producing virus. Assuming that the major protective effect of CTL is cytolytic, we demonstrate that mathematical models with an eclipse phase account for the data when the killing is fast and when it varies over the life cycle of infected cells. Considering the steady state corresponding to the chronic phase of the infection, we find that the rate of immune escape and the rate at which the viral load increases following CD8+ T cell depletion should reflect the viral replication rate, ρ. A meta-analysis of previous data shows that viral replication rates during chronic infection vary between 0.5 ≤ ρ ≤ 1 day−1. Balancing such fast viral replication requires killing rates that are several times larger than ρ, implying that most productively infected cells would die by cytolytic effects. IMPORTANCE Most current data suggest that cytotoxic T cells (CTL) mediate their control of human immunodeficiency virus type 1 (HIV-1) infection by nonlytic mechanisms; i.e., the data suggest that CTL hardly kill. This interpretation of these data has been based upon the general mathematical model for HIV infection. Because this model ignores the eclipse phase between the infection of a target cell and the start of viral production by that cell, we reanalyze the same data sets with novel models that do account for the eclipse phase. We find that the data are perfectly consistent with lytic control by CTL and predict that most productively infected cells are killed by CTL. Because the killing rate should balance the viral replication rate, we estimate both parameters from a large set of published experiments in which CD8+ T cells were depleted in simian immunodeficiency virus (SIV)-infected monkeys. This confirms that the killing rate can be much faster than is currently appreciated.
Collapse
|
359
|
Ishii H, Matsuoka S, Nomura T, Nakamura M, Shiino T, Sato Y, Iwata-Yoshikawa N, Hasegawa H, Mizuta K, Sakawaki H, Miura T, Koyanagi Y, Naruse TK, Kimura A, Matano T. Association of lymph-node antigens with lower Gag-specific central-memory and higher Env-specific effector-memory CD8(+) T-cell frequencies in a macaque AIDS model. Sci Rep 2016; 6:30153. [PMID: 27452272 PMCID: PMC4958968 DOI: 10.1038/srep30153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/27/2016] [Indexed: 01/17/2023] Open
Abstract
Virus-specific CD8+ T cells exert strong suppressive pressure on human/simian immunodeficiency virus (HIV/SIV) replication. These responses have been intensively examined in peripheral blood mononuclear cells (PBMCs) but not fully analyzed in lymph nodes (LNs), where interaction between CD8+ T cells and HIV/SIV-infected cells occurs. Here, we investigated target antigen specificity of CD8+ T cells in LNs in a macaque AIDS model. Analysis of virus antigen-specific CD8+ T-cell responses in the inguinal LNs obtained from twenty rhesus macaques in the chronic phase of SIV infection showed an inverse correlation between viral loads and frequencies of CD8+ T cells with CD28+ CD95+ central memory phenotype targeting the N-terminal half of SIV core antigen (Gag-N). In contrast, analysis of LNs but not PBMCs revealed a positive correlation between viral loads and frequencies of CD8+ T cells with CD28−CD95+ effector memory phenotype targeting the N-terminal half of SIV envelope (Env-N), soluble antigen. Indeed, LNs with detectable SIV capsid p27 antigen in the germinal center exhibited significantly lower Gag-N-specific CD28+ CD95+ CD8+ T-cell and higher Env-N-specific CD28−CD95+ CD8+ T-cell responses than those without detectable p27. These results imply that core and envelope antigen-specific CD8+ T cells show different patterns of interactions with HIV/SIV-infected cells.
Collapse
Affiliation(s)
- Hiroshi Ishii
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Saori Matsuoka
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Takushi Nomura
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,Center for AIDS Research, Kumamoto University, Tokyo 162-8640, Japan
| | - Midori Nakamura
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Teiichiro Shiino
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kazuta Mizuta
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Hiromi Sakawaki
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Tomoyuki Miura
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshio Koyanagi
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Taeko K Naruse
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
360
|
Boritz EA, Darko S, Swaszek L, Wolf G, Wells D, Wu X, Henry AR, Laboune F, Hu J, Ambrozak D, Hughes MS, Hoh R, Casazza JP, Vostal A, Bunis D, Nganou-Makamdop K, Lee JS, Migueles SA, Koup RA, Connors M, Moir S, Schacker T, Maldarelli F, Hughes SH, Deeks SG, Douek DC. Multiple Origins of Virus Persistence during Natural Control of HIV Infection. Cell 2016; 166:1004-1015. [PMID: 27453467 DOI: 10.1016/j.cell.2016.06.039] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/09/2016] [Accepted: 06/20/2016] [Indexed: 10/21/2022]
Abstract
Targeted HIV cure strategies require definition of the mechanisms that maintain the virus. Here, we tracked HIV replication and the persistence of infected CD4 T cells in individuals with natural virologic control by sequencing viruses, T cell receptor genes, HIV integration sites, and cellular transcriptomes. Our results revealed three mechanisms of HIV persistence operating within distinct anatomic and functional compartments. In lymph node, we detected viruses with genetic and transcriptional attributes of active replication in both T follicular helper (TFH) cells and non-TFH memory cells. In blood, we detected inducible proviruses of archival origin among highly differentiated, clonally expanded cells. Linking the lymph node and blood was a small population of circulating cells harboring inducible proviruses of recent origin. Thus, HIV replication in lymphoid tissue, clonal expansion of infected cells, and recirculation of recently infected cells act together to maintain the virus in HIV controllers despite effective antiviral immunity.
Collapse
Affiliation(s)
- Eli A Boritz
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Samuel Darko
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Luke Swaszek
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Gideon Wolf
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - David Wells
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Xiaolin Wu
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Amy R Henry
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Farida Laboune
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Jianfei Hu
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - David Ambrozak
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Marybeth S Hughes
- Thoracic and Gastrointestinal Oncology Branch, NCI, NIH, Bethesda, MD 20892, USA
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Joseph P Casazza
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Alexander Vostal
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Daniel Bunis
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - James S Lee
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Mark Connors
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD 20892, USA
| | - Susan Moir
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD 20892, USA
| | - Timothy Schacker
- Program in HIV Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA
| | - Stephen H Hughes
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
361
|
Abstract
Understanding where and how the HIV latent reservoir persists is essential for developing rational HIV cure strategies. In a recent paper in Nature, Lorenzo-Redondo et al. (2016) demonstrate that HIV persists and actively evolves within lymph nodes due to low antiretroviral drug penetration, revealing the need to target these drug-privileged sites.
Collapse
Affiliation(s)
- Anna Licht
- The Ragon Institute of MGH, MIT & Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Galit Alter
- The Ragon Institute of MGH, MIT & Harvard, 400 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
362
|
Anderson JL, Mota TM, Evans VA, Kumar N, Rezaei SD, Cheong K, Solomon A, Wightman F, Cameron PU, Lewin SR. Understanding Factors That Modulate the Establishment of HIV Latency in Resting CD4+ T-Cells In Vitro. PLoS One 2016; 11:e0158778. [PMID: 27383184 PMCID: PMC4934909 DOI: 10.1371/journal.pone.0158778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/21/2016] [Indexed: 11/18/2022] Open
Abstract
Developing robust in vitro models of HIV latency is needed to better understand how latency is established, maintained and reversed. In this study, we examined the effects of donor variability, HIV titre and co-receptor usage on establishing HIV latency in vitro using two models of HIV latency. Using the CCL19 model of HIV latency, we found that in up to 50% of donors, CCL19 enhanced latent infection of resting CD4+ T-cells by CXCR4-tropic HIV in the presence of low dose IL-2. Increasing the infectious titre of CXCR4-tropic HIV increased both productive and latent infection of resting CD4+ T-cells. In a different model where myeloid dendritic cells (mDC) were co-cultured with resting CD4+ T-cells, we observed a higher frequency of latently infected cells in vitro than CCL19-treated or unstimulated CD4+ T-cells in the presence of low dose IL-2. In the DC-T-cell model, latency was established with both CCR5- and CXCR4-tropic virus but higher titres of CCR5-tropic virus was required in most donors. The establishment of latency in vitro through direct infection of resting CD4+ T-cells is significantly enhanced by CCL19 and mDC, but the efficiency is dependent on virus titre, co-receptor usage and there is significant donor variability.
Collapse
Affiliation(s)
- Jenny L Anderson
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Talia M Mota
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Vanessa A Evans
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nitasha Kumar
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Simin D Rezaei
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Karey Cheong
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ajantha Solomon
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Fiona Wightman
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul U Cameron
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Sharon R Lewin
- Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
363
|
Abstract
PURPOSE OF REVIEW The introduction of effective antiretroviral therapy (ART) has transformed HIV infection from a deadly to a chronic infection. Despite its successes in reducing mortality, ART fails to cure HIV allowing HIV to persist in vivo. HIV persistence under ART is thought to be mediated by a combination of latent infection of long-lived cells, homeostatic proliferation of latently infected cells, anatomic sanctuaries, and low-level virus replication. To understand the contribution of specific cell types and anatomic sites to virus persistence in vivo animal models are necessary. RECENT FINDINGS The advancements in ART and our understanding of animal models have facilitated the development of models of HIV persistence in nonhuman primates and mice. Simian immunodeficiency virus (SIV) or simian/HIV infection (SHIV) of rhesus and pigtail macaques followed by effective ART represents the most faithful animal model of HIV persistence. HIV infection of humanized mice also provides a useful model for answering specific questions regarding virus persistence in a uniquely mutable system. SUMMARY In this review, we describe the most recent findings using animal models of HIV persistence. We will first describe the important aspects of HIV infection that SIV/SHIV infection of nonhuman primates are able to recapitulate, then we will discuss some recent studies that have used these models to understand viral persistence.
Collapse
|
364
|
Persistence of integrated HIV DNA in CXCR3 + CCR6 + memory CD4+ T cells in HIV-infected individuals on antiretroviral therapy. AIDS 2016; 30:1511-20. [PMID: 26807971 DOI: 10.1097/qad.0000000000001029] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND HIV latent infection can be established in vitro by treating resting CD4 T cells with chemokines that bind to chemokine receptors (CKR), CCR7, CXCR3, and CCR6, highly expressed on T cells. OBJECTIVE To determine if CKR identify CD4 T cells enriched for HIV in HIV-infected individuals receiving suppressive antiretroviral therapy (ART). DESIGN A cross-sectional study of CKR expression and HIV persistence in blood from HIV-infected individuals on suppressive ART for more than 3 years (n = 48). A subset of 20 individuals underwent leukapheresis and sorting of specific CD4 T-cell subsets. METHODS We used flow cytometry to quantify CCR5, CCR6, CXCR3, and CXCR5 expression on CD4 T cells. HIV persistence was quantified using real-time Polymerase Chain Reaction to detect total, integrated HIV DNA, 2-long terminal repeat circles and cell-associated unspliced (CA-US) HIV RNA in total CD4 T cells from blood or sorted T-cell subsets. Associations between CKR and HIV persistence in CD4 T cells in blood were determined using regression models and adjusted for current and nadir CD4 T-cell counts. RESULTS The frequency of cells harbouring integrated HIV DNA was inversely associated with current CD4 T-cell count and positively associated with CCR5+ CD4 T cells, CXCR3+CCR6+ and CXCR3+CCR6- expression on total memory CD4 T cells (P < 0.001, 0.048, 0.015, and 0.016, respectively). CXCR3+CCR6+ CM CD4 T cells contained the highest amount of integrated HIV DNA and lowest ratio of CA-US HIV RNA to DNA compared to all T-cell subsets examined. CONCLUSION CXCR3 and CCR6 coexpression defines a subset of CD4 T cells that are preferentially enriched for HIV DNA in HIV-infected individuals on ART.
Collapse
|
365
|
Polizzotto MN, Chen G, Tressler RL, Godfrey C. Leveraging Cancer Therapeutics for the HIV Cure Agenda: Current Status and Future Directions. Drugs 2016. [PMID: 26224205 DOI: 10.1007/s40265-015-0426-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite effective antiretroviral therapy (ART) and undetectable HIV RNA in the plasma, latent replication-competent HIV persists indefinitely in long-lived cells. Cessation of ART results in rebound of HIV from these persistent reservoirs. While this was thought to be an insurmountable obstacle to viral eradication, recent cases suggest otherwise. To date one patient has been "cured" of HIV and several others have been able to interrupt ART without viral rebound for prolonged periods. These events have sparked renewed interest in developing strategies that will allow eradication of HIV in infected individuals. We review the current knowledge of HIV latency and the viral reservoir, describe the potential utility of emerging cancer therapeutics in HIV cure research with an emphasis on pathways implicated in reservoir persistence, and outline opportunities and challenges in the context of the current clinical trial and regulatory environment.
Collapse
Affiliation(s)
- Mark N Polizzotto
- HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, 5601 Fishers Lane, Bethesda, MD, 20892, USA,
| | | | | | | |
Collapse
|
366
|
Kimata JT, Rice AP, Wang J. Challenges and strategies for the eradication of the HIV reservoir. Curr Opin Immunol 2016; 42:65-70. [PMID: 27288651 DOI: 10.1016/j.coi.2016.05.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/29/2016] [Indexed: 12/31/2022]
Abstract
Despite the success of highly active antiretroviral therapy (HAART) for inhibiting HIV replication and improving clinical outcomes, it fails to cure infection due to the existence of a stable latent proviral reservoir in memory CD4+ T cells. Because of the longevity of these cells harboring transcriptionally silent proviruses, devising strategies to induce viral gene expression so the host immune response can mediate clearance of the infected cells or the cells can undergo virus-induced cell death, has been of considerable recent interest. Here, we review current knowledge of latency, and the challenges to virus induction and eradication. Novel strategies to reactivate HIV reservoirs more effectively, in combination with immunotherapy, could lead to better clearance of the latent HIV reservoir.
Collapse
Affiliation(s)
- Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.
| | - Andrew P Rice
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Jin Wang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
367
|
Elimination of Latently HIV-infected Cells from Antiretroviral Therapy-suppressed Subjects by Engineered Immune-mobilizing T-cell Receptors. Mol Ther 2016; 24:1913-1925. [PMID: 27401039 PMCID: PMC5154472 DOI: 10.1038/mt.2016.114] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 05/24/2016] [Indexed: 01/06/2023] Open
Abstract
Persistence of human immunodeficiency virus (HIV) in a latent state in long-lived CD4+ T-cells is a major barrier to eradication. Latency-reversing agents that induce direct or immune-mediated cell death upon reactivation of HIV are a possible solution. However, clearance of reactivated cells may require immunotherapeutic agents that are fine-tuned to detect viral antigens when expressed at low levels. We tested the antiviral efficacy of immune-mobilizing monoclonal T-cell receptors against viruses (ImmTAVs), bispecific molecules that redirect CD8+ T-cells to kill HIV-infected CD4+ T-cells. T-cell receptors specific for an immunodominant Gag epitope, SL9, and its escape variants were engineered to achieve supraphysiological affinity and fused to a humanised CD3-specific single chain antibody fragment. Ex vivo polyclonal CD8+ T-cells were efficiently redirected by immune-mobilising monoclonal T-cell receptors against viruses to eliminate CD4+ T-cells from human histocompatibility leukocyte antigen (HLA)-A*0201-positive antiretroviral therapy-treated patients after reactivation of inducible HIV in vitro. The efficiency of infected cell elimination correlated with HIV Gag expression. Immune-mobilising monoclonal T-cell receptors against viruses have potential as a therapy to facilitate clearance of reactivated HIV reservoir cells.
Collapse
|
368
|
Banga R, Procopio FA, Noto A, Pollakis G, Cavassini M, Ohmiti K, Corpataux JM, de Leval L, Pantaleo G, Perreau M. PD-1+ and follicular helper T cells are responsible for persistent HIV-1 transcription in treated aviremic individuals. Nat Med 2016; 22:754-61. [DOI: 10.1038/nm.4113] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/26/2016] [Indexed: 12/13/2022]
|
369
|
Affiliation(s)
- Carola G. Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia;
| | - Michelle A. Linterman
- Lymphocyte Signalling and Development Institute Strategic Programme, Babraham Institute, Cambridge CB22 3AT, United Kingdom;
| | - Di Yu
- Laboratory for Molecular Immunomodulation, Department of Biochemistry and Molecular Biology, and Center for Inflammatory Diseases, Monash University, Melbourne, Victoria 3800, Australia;
| | - Ian C.M. MacLennan
- School of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
370
|
Stevenson M. CROI 2016: Basic Science Review. TOPICS IN ANTIVIRAL MEDICINE 2016; 24:4-9. [PMID: 27398858 PMCID: PMC6148922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 06/06/2023]
Abstract
The 2016 Conference on Retroviruses and Opportunistic Infections continued to maintain balance in the representation of different areas of research related to HIV/AIDS. The basic science category encompasses research on viral reservoirs and HIV cure, on cellular factors regulating the interplay between virus and host, and on factors that influence viral pathogenicity. Basic research on factors that influence the interaction between the virus and the host cell continues to unearth surprises with the identification of a new host antiviral factor. Further, research into the mechanisms of viral persistence reveals that there is much to learn about how HIV-1 is able to persist in the face of antiviral suppression.
Collapse
|
371
|
Luo S, Perelson AS. The challenges of modelling antibody repertoire dynamics in HIV infection. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0247. [PMID: 26194760 DOI: 10.1098/rstb.2014.0247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Antibody affinity maturation by somatic hypermutation of B-cell immunoglobulin variable region genes has been studied for decades in various model systems using well-defined antigens. While much is known about the molecular details of the process, our understanding of the selective forces that generate affinity maturation are less well developed, particularly in the case of a co-evolving pathogen such as HIV. Despite this gap in understanding, high-throughput antibody sequence data are increasingly being collected to investigate the evolutionary trajectories of antibody lineages in HIV-infected individuals. Here, we review what is known in controlled experimental systems about the mechanisms underlying antibody selection and compare this to the observed temporal patterns of antibody evolution in HIV infection. We describe how our current understanding of antibody selection mechanisms leaves questions about antibody dynamics in HIV infection unanswered. Without a mechanistic understanding of antibody selection in the context of a co-evolving viral population, modelling and analysis of antibody sequences in HIV-infected individuals will be limited in their interpretation and predictive ability.
Collapse
Affiliation(s)
- Shishi Luo
- Department of Electrical Engineering and Computer Science, UC Berkeley, Berkeley, CA 94110, USA Department of Statistics, UC Berkeley, Berkeley, CA 94110, USA
| | - Alan S Perelson
- Los Alamos National Laboratory, Theoretical Biology and Biophysics Group, Los Alamos, NM 87545, USA
| |
Collapse
|
372
|
Elevation and persistence of CD8 T-cells in HIV infection: the Achilles heel in the ART era. J Int AIDS Soc 2016; 19:20697. [PMID: 26945343 PMCID: PMC4779330 DOI: 10.7448/ias.19.1.20697] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 01/15/2016] [Accepted: 01/28/2016] [Indexed: 12/18/2022] Open
Abstract
Introduction HIV infection leads to a disturbed T-cell homeostasis, featured by a depletion of CD4 T-cells and a persistent elevation of CD8 T-cells over disease progression. Most effort of managing HIV infection has been focused on CD4 T-cell recovery, while changes in the CD8 compartment were relatively underappreciated in the past. Methods A comprehensive literature review of publications in English language was conducted using major electronic databases. Our search was focused on factors contributing to CD8 T-cell dynamics in HIV infection and following antiretroviral therapy (ART). Discussion Normalization of CD8 counts is seldom observed even with optimal CD4 recovery following long-term treatment. Initiation of ART in primary HIV infection leads to enhanced normalization of CD8 count compared with long-term ART initiated in chronic infection. Importantly, such CD8 elevation in treated HIV infection is associated with an increased risk of inflammatory non-AIDS-related clinical events independent of CD4 T-cell recovery. The mechanisms underlying CD8 persistence remain largely unknown, which may include bystander activation, exhaustion and immunosenescence of CD8 T-cells. The information provided herein will lead to a better understanding of factors associated with CD8 persistence and contribute to the development of strategies aiming at CD8 normalization. Conclusions Persistence of CD8 T-cell elevation in treated HIV-infected patients is associated with an increased risk of non-AIDS-related events. Now that advances in ART have led to decreased AIDS-related opportunistic diseases, more attention has been focused on reducing non-AIDS events and normalizing persistent CD8 T-cell elevation.
Collapse
|
373
|
Miles B, Connick E. TFH in HIV Latency and as Sources of Replication-Competent Virus. Trends Microbiol 2016; 24:338-344. [PMID: 26947191 DOI: 10.1016/j.tim.2016.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 11/16/2022]
Abstract
During untreated disease, HIV replication is concentrated within T follicular helper cells (TFH). Heightened permissiveness, the presence of highly infectious virions on follicular dendritic cells (FDCs), low frequencies of virus-specific cytotoxic T lymphocytes (CTLs) in B cell follicles, expansions in TFH, and TFH dysfunction, all likely promote replication in TFH. Limited data suggest that memory TFH play a role in the latent or subclinical reservoir of HIV during antiretroviral therapy (ART), potentially for many of the same reasons. A better understanding of the role of memory TFH and FDC-bound virions in promoting recrudescent viremia in the setting of ART cessation is essential. Studies that target follicular virus reservoirs are needed to determine their role in HIV latency and to suggest successful cure strategies.
Collapse
Affiliation(s)
- Brodie Miles
- Division of Infectious Diseases, University of Colorado Denver, Aurora CO 80045, USA
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Colorado Denver, Aurora CO 80045, USA.
| |
Collapse
|
374
|
Massanella M, Fromentin R, Chomont N. Residual inflammation and viral reservoirs: alliance against an HIV cure. Curr Opin HIV AIDS 2016; 11:234-41. [PMID: 26575148 PMCID: PMC4743501 DOI: 10.1097/coh.0000000000000230] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW HIV persists in cellular and anatomical reservoirs during antiretroviral therapy (ART). Viral persistence is ensured by a variety of mechanisms including ongoing viral replication and proliferation of latently infected cells. In this review, we summarize recent findings establishing a link between the unresolved levels of inflammation observed in virally suppressed individuals on ART and the mechanisms responsible for HIV persistence. RECENT FINDINGS Residual levels of viral replication during ART are associated with persistent low levels of immune activation, suggesting that unresolved inflammation can promote the replenishment of the HIV reservoir in tissues. In addition, the recent findings that the latent HIV reservoir is maintained by continuous proliferation of latently infected cells provide another mechanism by which residual inflammation could contribute to HIV persistence. SUMMARY Residual inflammation during ART is likely to be a critical parameter contributing to HIV persistence. Therefore, reducing inflammation may be an efficient way to interfere with the maintenance of the HIV reservoir in virally suppressed individuals on ART.
Collapse
Affiliation(s)
- Marta Massanella
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| | - Rémi Fromentin
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| | - Nicolas Chomont
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| |
Collapse
|
375
|
Kohler SL, Pham MN, Folkvord JM, Arends T, Miller SM, Miles B, Meditz AL, McCarter M, Levy DN, Connick E. Germinal Center T Follicular Helper Cells Are Highly Permissive to HIV-1 and Alter Their Phenotype during Virus Replication. THE JOURNAL OF IMMUNOLOGY 2016; 196:2711-22. [PMID: 26873986 DOI: 10.4049/jimmunol.1502174] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/18/2016] [Indexed: 01/13/2023]
Abstract
HIV-1 replication is concentrated within CD4(+) T cells in B cell follicles of secondary lymphoid tissues during asymptomatic disease. Limited data suggest that a subset of T follicular helper cells (TFH) within germinal centers (GC) is highly permissive to HIV-1. Whether GC TFH are the major HIV-1 virus-producing cells in vivo has not been established. In this study, we investigated TFH permissivity to HIV-1 ex vivo by spinoculating and culturing tonsil cells with HIV-1 GFP reporter viruses. Using flow cytometry, higher percentages of GC TFH (CXCR5(high)PD-1(high)) and CXCR5(+)programmed cell death-1 (PD-1)(low) cells were GFP(+) than non-GC TFH (CXCR5(+)PD-1(intermediate)) or extrafollicular (EF) (CXCR5(-)) cells. When sorted prior to spinoculation, however, GC TFH were substantially more permissive than CXCR5(+)PD-1(low) or EF cells, suggesting that many GC TFH transition to a CXCR5(+)PD-1(low) phenotype during productive infection. In situ hybridization on inguinal lymph node sections from untreated HIV-1-infected individuals without AIDS revealed higher frequencies of HIV-1 RNA(+) cells in GC than non-GC regions of follicle or EF regions. Superinfection of HIV-1-infected individuals' lymph node cells with GFP reporter virus confirmed the permissivity of follicular cells ex vivo. Lymph node immunostaining revealed 96% of CXCR5(+)CD4(+) cells were located in follicles. Within sorted lymph node cells from four HIV-infected individuals, CXCR5(+) subsets harbored 11-66-fold more HIV-1 RNA than CXCR5(-) subsets, as determined by RT PCR. Thus, GC TFH are highly permissive to HIV-1, but downregulate PD-1 and, to a lesser extent, CXCR5 during HIV-1 replication. These data further implicate GC TFH as the major HIV-1-producing cells in chronic asymptomatic HIV-1 infection.
Collapse
Affiliation(s)
- Stephanie L Kohler
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael N Pham
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Joy M Folkvord
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Tessa Arends
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Shannon M Miller
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Brodie Miles
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Amie L Meditz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Martin McCarter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and
| | - David N Levy
- College of Dentistry, New York University, New York, NY 10010
| | - Elizabeth Connick
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
| |
Collapse
|
376
|
Denton PW, Søgaard OS, Tolstrup M. Using animal models to overcome temporal, spatial and combinatorial challenges in HIV persistence research. J Transl Med 2016; 14:44. [PMID: 26861779 PMCID: PMC4746773 DOI: 10.1186/s12967-016-0807-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/29/2016] [Indexed: 12/03/2022] Open
Abstract
Research challenges associated with understanding HIV persistence during antiretroviral therapy can be categorized as temporal, spatial and combinatorial. Temporal research challenges relate to the timing of events during establishment and maintenance of HIV persistence. Spatial research challenges regard the anatomical locations and cell subsets that harbor persistent HIV. Combinatorial research challenges pertain to the order of administration, timing of administration and specific combinations of compounds to be administered during HIV eradication therapy. Overcoming these challenges will improve our understanding of HIV persistence and move the field closer to achieving eradication of persistent HIV. Given that humanized mice and non-human primate HIV models permit rigorous control of experimental conditions, these models have been used extensively as in vivo research platforms for directly addressing these research challenges. The aim of this manuscript is to provide a comprehensive review of these recent translational advances made in animal models of HIV persistence.
Collapse
Affiliation(s)
- Paul W Denton
- Institute of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark. .,Department of Infectious Diseases, Aarhus University Hospital, Skejby, Aarhus, Denmark. .,Aarhus Institute for Advanced Studies, Aarhus University, Aarhus, Denmark.
| | - Ole S Søgaard
- Institute of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark. .,Department of Infectious Diseases, Aarhus University Hospital, Skejby, Aarhus, Denmark.
| | - Martin Tolstrup
- Institute of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark. .,Department of Infectious Diseases, Aarhus University Hospital, Skejby, Aarhus, Denmark.
| |
Collapse
|
377
|
Siliciano JD, Siliciano RF. Recent developments in the effort to cure HIV infection: going beyond N = 1. J Clin Invest 2016; 126:409-14. [PMID: 26829622 DOI: 10.1172/jci86047] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Combination antiretroviral therapy (ART) can suppress plasma HIV to undetectable levels, allowing HIV-infected individuals who are treated early a nearly normal life span. Despite the clear ability of ART to prevent morbidity and mortality, it is not curative. Even in individuals who have full suppression of viral replication on ART, there are resting memory CD4+ T cells that harbor stably integrated HIV genomes, which are capable of producing infectious virus upon T cell activation. This latent viral reservoir is considered the primary obstacle to the development of an HIV cure, and recent efforts in multiple areas of HIV research have been brought to bear on the development of strategies to eradicate or develop a functional cure for HIV. Reviews in this series detail progress in our understanding of the molecular and cellular mechanisms of viral latency, efforts to accurately assess the size and composition of the latent reservoir, the characterization and development of HIV-targeted broadly neutralizing antibodies and cytolytic T lymphocytes, and animal models for the study HIV latency and therapeutic strategies.
Collapse
|
378
|
Vargas-Inchaustegui DA, Demers A, Shaw JM, Kang G, Ball D, Tuero I, Musich T, Mohanram V, Demberg T, Karpova TS, Li Q, Robert-Guroff M. Vaccine Induction of Lymph Node-Resident Simian Immunodeficiency Virus Env-Specific T Follicular Helper Cells in Rhesus Macaques. THE JOURNAL OF IMMUNOLOGY 2016; 196:1700-10. [PMID: 26773147 DOI: 10.4049/jimmunol.1502137] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/10/2015] [Indexed: 12/24/2022]
Abstract
Measurement of Ag-specific T follicular helper (TFH) cell activity in rhesus macaques has not previously been reported. Given that rhesus macaques are the animal model of choice for evaluating protective efficacy of HIV/SIV vaccine candidates and that TFH cells play a pivotal role in aiding B cell maturation, quantifying vaccine induction of HIV/SIV-specific TFH cells would greatly benefit vaccine development. In this study, we quantified SIV Env-specific IL-21-producing TFH cells for the first time, to our knowledge, in a nonhuman primate vaccine study. Macaques were primed twice mucosally with adenovirus 5 host range mutant recombinants encoding SIV Env, Rev, Gag, and Nef followed by two i.m. boosts with monomeric SIV gp120 or oligomeric SIV gp140 proteins. At 2 wk after the second protein boost, we obtained lymph node biopsy specimens and quantified the frequency of total and SIV Env-specific IL-21(+) TFH cells and total germinal center B cells, the size and number of germinal centers, and the frequency of SIV-specific Ab-secreting cells in B cell zones. Multiple correlation analyses established the importance of TFH for development of B cell responses in systemic and mucosally localized compartments, including blood, bone marrow, and rectum. Our results suggest that the SIV-specific TFH cells, initially induced by replicating adenovirus-recombinant priming, are long lived. The multiple correlations of SIV Env-specific TFH cells with systemic and mucosal SIV-specific B cell responses indicate that this cell population should be further investigated in HIV vaccine development as a novel correlate of immunity.
Collapse
Affiliation(s)
- Diego A Vargas-Inchaustegui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| | - Andrew Demers
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - Julia M Shaw
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Guobin Kang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - David Ball
- Center for Cancer Research Core Fluorescence Imaging Facility, Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Iskra Tuero
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Thomas Musich
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Venkatramanan Mohanram
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Thorsten Demberg
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tatiana S Karpova
- Center for Cancer Research Core Fluorescence Imaging Facility, Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - Marjorie Robert-Guroff
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
379
|
Halper-Stromberg A, Nussenzweig MC. Towards HIV-1 remission: potential roles for broadly neutralizing antibodies. J Clin Invest 2016; 126:415-23. [PMID: 26752643 DOI: 10.1172/jci80561] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Current antiretroviral drug therapies do not cure HIV-1 because they do not eliminate a pool of long-lived cells harboring immunologically silent but replication-competent proviruses - termed the latent reservoir. Eliminating this reservoir and stimulating the immune response to control infection in the absence of therapy remain important but unsolved goals of HIV-1 cure research. Recently discovered broadly neutralizing antibodies (bNAbs) exhibit remarkable breadth and potency in their ability to neutralize HIV-1 in vitro, and recent studies have demonstrated new therapeutic applications for passively administered bNAbs in vivo. This Review discusses the roles bNAbs might play in HIV-1 treatment regimens, including prevention, therapy, and cure.
Collapse
|
380
|
Towards an HIV cure based on targeted killing of infected cells: different approaches against acute versus chronic infection. Curr Opin HIV AIDS 2016; 10:207-13. [PMID: 25710815 DOI: 10.1097/coh.0000000000000151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Current regimens of combination antiretroviral therapy (cART) offer effective control of HIV infection, with maintenance of immune health and near-normal life expectancy. What will it take to progress beyond the status quo, whereby infectious virus can be eradicated (a 'sterilizing cure') or fully controlled without the need for ongoing cART (a 'functional cure')? RECENT FINDINGS On the basis of therapeutic advances in the cancer field, we propose that targeted cytotoxic therapy to kill HIV-infected cells represents a logical complement to cART for achieving an HIV cure. This concept is based on the fact that cART effectively blocks replication of the virus, but does not eliminate cells that are already infected; targeted cytotoxic therapy would contribute precisely this missing component. We suggest that different modalities are suited for curing primary acute versus established chronic infection. For acute infection, relatively short-acting potent agents such as recombinant immunotoxins might prove sufficient for HIV eradication, whereas for chronic infection, a long-lasting (lifelong?) modality is required to maintain full virus control, as might be achieved with genetically modified autologous T cells. SUMMARY We present perspectives for complementing cART with targeted cytotoxic therapy, whereby HIV infection is either eradicated or fully controlled, thereby eliminating the need for lifelong cART.
Collapse
|
381
|
Abstract
After the success of combination antiretroviral therapy (cART) to treat HIV infection, the next great frontier is to cure infected persons, a formidable challenge. HIV persists in a quiescent state in resting CD4+ T cells, where the replicative enzymes targeted by cART are not active. Although low levels of HIV transcripts are detectable in these resting cells, little to no viral protein is produced, rendering this reservoir difficult to detect by the host CD8+ T cell response. However, recent advances suggest that this state of latency might be pharmacologically reversed, resulting in viral protein expression without the adverse effects of massive cellular activation. Emerging data suggest that with this approach, infected cells will not die of viral cytopathic effects, but might be eliminated if HIV-specific CD8+ T cells can be effectively harnessed. Here, we address the antiviral properties of HIV-specific CD8+ T cells and how these cells might be harnessed to greater effect toward achieving viral eradication or a functional cure.
Collapse
|
382
|
Deleage C, Wietgrefe SW, Del Prete G, Morcock DR, Hao XP, Piatak M, Bess J, Anderson JL, Perkey KE, Reilly C, McCune JM, Haase AT, Lifson JD, Schacker TW, Estes JD. Defining HIV and SIV Reservoirs in Lymphoid Tissues. Pathog Immun 2016; 1:68-106. [PMID: 27430032 PMCID: PMC4943335 DOI: 10.20411/pai.v1i1.100] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A primary obstacle to an HIV-1 cure is long-lived viral reservoirs, which must be eliminated or greatly reduced. Cure strategies have largely focused on monitoring changes in T cell reservoirs in peripheral blood (PB), even though the lymphoid tissues (LT) are primary sites for viral persistence. To track and discriminate viral reservoirs within tissue compartments we developed a specific and sensitive next-generation in situ hybridization approach to detect vRNA, including vRNA+ cells and viral particles ("RNAscope"), vDNA+ cells ("DNAscope") and combined vRNA and vDNA with immunohistochemistry to detect and phenotype active and latently infected cells in the same tissue section. RNAscope is highly sensitive with greater speed of analysis compared to traditional in situ hybridization. The highly sensitive and specific DNAscope detected SIV/HIV vDNA+ cells, including duplexed detection of vDNA and vRNA or immunophenotypic markers in the same section. Analysis of LT samples from macaques prior to and during combination antiretroviral therapy demonstrated that B cell follicles are an important anatomical compartment for both latent and active viral persistence during treatment. These new tools should allow new insights into viral reservoir biology and evaluation of cure strategies.
Collapse
Affiliation(s)
- Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Stephen W. Wietgrefe
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Gregory Del Prete
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - David R. Morcock
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Xing Pei Hao
- Pathology and Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
- Deceased 19 September 2014
| | - Julian Bess
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jodi L. Anderson
- Department of Medicine. Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Katherine E. Perkey
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Cavan Reilly
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| | - Ashley T. Haase
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Timothy W. Schacker
- Department of Medicine. Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
383
|
Margolis DM, Salzwedel K, Chomont N, Psomas C, Routy JP, Poli G, Lafeuillade A. Highlights from the Seventh International Workshop on HIV Persistence during Therapy, 8–11 December 2015, Miami, Florida, USA. J Virus Erad 2016. [DOI: 10.1016/s2055-6640(20)30684-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
384
|
Margolis DM, Salzwedel K, Chomont N, Psomas C, Routy JP, Poli G, Lafeuillade A. Highlights from the Seventh International Workshop on HIV Persistence during Therapy, 8-11 December 2015, Miami, Florida, USA. J Virus Erad 2016; 2:57-65. [PMID: 27482437 PMCID: PMC4946700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
Over 4 days, more than 270 scientists involved in HIV persistence research convened to share their data and discuss future avenues to control HIV without continuous antiretroviral therapy. This 7(th) International Workshop on HIV Persistence followed the format of the preceding conferences but more time was given for discussing abstracts submitted by the participants and selected by the Steering and Scientific Committees. The topic of the workshop is HIV persistence: consequently, issues of HIV reservoirs and HIV cure are also addressed. In this article we report as closely as possible what was discussed. However, owing to length constraints, not everything is reported here but all the Workshop abstracts can be found online (www.viruseradication.com).
Collapse
Affiliation(s)
| | - Karl Salzwedel
- National Institute of Allergy and Infectious Diseases, Bethesda, USA
| | | | | | | | - Guido Poli
- San Raffaele Scientific Institute, Milano, Italy
| | | |
Collapse
|
385
|
Peripheral T Follicular Helper Cells Are the Major HIV Reservoir within Central Memory CD4 T Cells in Peripheral Blood from Chronically HIV-Infected Individuals on Combination Antiretroviral Therapy. J Virol 2015; 90:2718-28. [PMID: 26676775 DOI: 10.1128/jvi.02883-15] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/10/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED In this study, we examined the peripheral blood (PB) central memory (TCM) CD4(+) T cell subsets designated peripheral T follicular helper cells (pTfh cells) and non-pTfh cells to assess HIV permissiveness and persistence. Purified pTfh and non-pTfh cells from healthy HIV-negative donors were tested for HIV permissiveness using green fluorescent protein (GFP)-expressing HIV-1NL4-3/Ba-L, followed by viral reactivation using beads coated with anti-CD3/anti-CD28 monoclonal antibodies. The role of pTfh cells in HIV persistence was analyzed in 12 chronically HIV-1 infected patients before and 48 weeks after initiation of raltegravir-containing combination antiretroviral therapy (cART). Total cellular HIV-1 DNA and episomes containing two copies of the viral long terminal repeat (2LTR circles) were analyzed in using droplet digital PCR in the purified pTfh and non-pTfh cells. Activation-inducible HIV p24 expression was determined by flow cytometry. Results indicate that pTfh cells, in particular PD1(+) pTfh cells, showed greater permissiveness for HIV infection than non-pTfh cells. At week 48 on cART, HIV DNA levels were unchanged from pre-cART levels, although a significant decrease in 2LTR circles was observed in both cell subsets. Inducible HIV p24 expression was higher in pTfh cells than in non-pTfh cells, with the highest frequencies in the PD1(+) CXCR3(-) pTfh cell subset. Frequencies of HLADR(+) CD38(+) activated CD4 T cells correlated with 2LTR circles in pTfh and non-pTfh cells at both time points and with p24(+) cells at entry. In conclusion, among CD4 TCM cells in PB of aviremic patients on cART, pTfh cells, in particular the PD1(+) CXCR3(-) subset, constitute a major HIV reservoir that is sustained by ongoing residual immune activation. The inducible HIV p24 assay is useful for monitoring HIV reservoirs in defined CD4 T cell subsets. IMPORTANCE Identification of the type and nature of the cellular compartments of circulating HIV reservoirs is important for targeting of HIV cure strategies. In lymph nodes (LN), a subset of CD4 T cells called T follicular helper (Tfh) cells are preferentially infected by HIV. Central memory (TCM) CD4 T cells are the major cellular reservoir for HIV in peripheral blood and contain a subset of CD4 TCM cells expressing chemokine receptor CXCR5 similar in function to LN Tfh cells termed peripheral Tfh (pTfh) cells. We found that the circulating pTfh cells are highly susceptible to HIV infection and that in HIV-infected patients, HIV persists in these cells following plasma virus suppression with potent cART. These pTfh cells, which constitute a subset of TCM CD4 T cells, can be readily monitored in peripheral blood to assess HIV persistence.
Collapse
|
386
|
Micci L, Ryan ES, Fromentin R, Bosinger SE, Harper JL, He T, Paganini S, Easley KA, Chahroudi A, Benne C, Gumber S, McGary CS, Rogers KA, Deleage C, Lucero C, Byrareddy SN, Apetrei C, Estes JD, Lifson JD, Piatak M, Chomont N, Villinger F, Silvestri G, Brenchley JM, Paiardini M. Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques. J Clin Invest 2015; 125:4497-513. [PMID: 26551680 PMCID: PMC4665780 DOI: 10.1172/jci81400] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 09/24/2015] [Indexed: 02/06/2023] Open
Abstract
Despite successful control of viremia, many HIV-infected individuals given antiretroviral therapy (ART) exhibit residual inflammation, which is associated with non-AIDS-related morbidity and mortality and may contribute to virus persistence during ART. Here, we investigated the effects of IL-21 administration on both inflammation and virus persistence in ART-treated, SIV-infected rhesus macaques (RMs). Compared with SIV-infected animals only given ART, SIV-infected RMs given both ART and IL-21 showed improved restoration of intestinal Th17 and Th22 cells and a more effective reduction of immune activation in blood and intestinal mucosa, with the latter maintained through 8 months after ART interruption. Additionally, IL-21, in combination with ART, was associated with reduced levels of SIV RNA in plasma and decreased CD4(+) T cell levels harboring replication-competent virus during ART. At the latest experimental time points, which were up to 8 months after ART interruption, plasma viremia and cell-associated SIV DNA levels remained substantially lower than those before ART initiation in IL-21-treated animals but not in controls. Together, these data suggest that IL-21 supplementation of ART reduces residual inflammation and virus persistence in a relevant model of lentiviral disease and warrants further investigation as a potential intervention for HIV infection.
Collapse
Affiliation(s)
- Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Emily S. Ryan
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rémi Fromentin
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, and Centre de Recherche du CHUM, Montreal, Quebec, Canada
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Yerkes Nonhuman Primate Genomics Core, Emory University, Atlanta, Georgia, USA
| | - Justin L. Harper
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tianyu He
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sara Paganini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kirk A. Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, and
| | - Ann Chahroudi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Clarisse Benne
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Colleen S. McGary
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kenneth A. Rogers
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Claire Deleage
- AIDS Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Carissa Lucero
- AIDS Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Siddappa N. Byrareddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jacob D. Estes
- AIDS Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Jeffrey D. Lifson
- AIDS Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Michael Piatak
- AIDS Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Nicolas Chomont
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, and Centre de Recherche du CHUM, Montreal, Quebec, Canada
| | - Francois Villinger
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jason M. Brenchley
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
387
|
Moukambi F, Rabezanahary H, Rodrigues V, Racine G, Robitaille L, Krust B, Andreani G, Soundaramourty C, Silvestre R, Laforge M, Estaquier J. Early Loss of Splenic Tfh Cells in SIV-Infected Rhesus Macaques. PLoS Pathog 2015; 11:e1005287. [PMID: 26640894 PMCID: PMC4671657 DOI: 10.1371/journal.ppat.1005287] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 10/28/2015] [Indexed: 11/28/2022] Open
Abstract
Follicular T helper cells (Tfh), a subset of CD4 T lymphocytes, provide crucial help to B cells in the production of antigen-specific antibodies. Although several studies have analyzed the dynamics of Tfh cells in peripheral blood and lymph nodes (LNs) during Aids, none has yet addressed the impact of SIV infection on the dynamics of Tfh cells in the spleen, the primary organ of B cell activation. We show here a significant decrease in splenic Tfh cells in SIVmac251-infected rhesus macaques (RMs) during the acute phase of infection, which persists thereafter. This profound loss is associated with lack of sustained expression of the Tfh-defining transcription factors, Bcl-6 and c-Maf but with higher expression of the repressors KLF2 and Foxo1. In this context of Tfh abortive differentiation and loss, we found decreased percentages of memory B cell subsets and lower titers of SIV-specific IgG. We further demonstrate a drastic remodeling of the lymphoid architecture of the spleen and LNs, which disrupts the crucial cell-cell interactions necessary to maintain memory B cells and Tfh cells. Finally, our data demonstrated the early infection of Tfh cells. Paradoxically, the frequencies of SIV DNA were higher in splenic Tfh cells of RMs progressing more slowly suggesting sanctuaries for SIV in the spleen. Our findings provide important information regarding the impact of HIV/SIV infection on Tfh cells, and provide new clues for future vaccine strategies.
Collapse
Affiliation(s)
- Félicien Moukambi
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medecine, Laval University, Québec, Québec, Canada
| | - Henintsoa Rabezanahary
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medecine, Laval University, Québec, Québec, Canada
| | - Vasco Rodrigues
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France
| | - Gina Racine
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medecine, Laval University, Québec, Québec, Canada
| | - Lynda Robitaille
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medecine, Laval University, Québec, Québec, Canada
| | - Bernard Krust
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France
| | - Guadalupe Andreani
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medecine, Laval University, Québec, Québec, Canada
| | | | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Mireille Laforge
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France
| | - Jérôme Estaquier
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medecine, Laval University, Québec, Québec, Canada
- CNRS FR3636, Faculty of Medecine des Saint-Pères, Paris Descartes University, Paris, France
| |
Collapse
|
388
|
Persistent Simian Immunodeficiency Virus Infection Drives Differentiation, Aberrant Accumulation, and Latent Infection of Germinal Center Follicular T Helper Cells. J Virol 2015; 90:1578-87. [PMID: 26608323 DOI: 10.1128/jvi.02471-15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/17/2015] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED CD4(+) follicular T helper (Tfh) cells play a prominent role in humoral immune responses, but the mechanisms of their accumulation and infection in AIDS remain unclear. Here we found that germinal center (GC) Tfh cells, defined here as CXCR5(+) PD-1(HIGH) CD4(+) T cells, do not express the HIV coreceptor CCR5 yet serve as a latent reservoir in GCs. With disease progression, an expansion of GC Tfh cells is accompanied by increases in dysfunctional CD8(+) T cells. In contrast, Tfh precursor (CXCR5(-) CD4(+) T) cells in lymph nodes do express CCR5 and differentiate into GC Tfh cells following interleukin-6 (IL-6) and IL-21 stimulation, and viral DNA is detectable in fully differentiated GC Tfh cells ex vivo. This suggests that SIV-infected GC Tfh cells may be derived from Tfh precursor cell subsets that become infected in marginal zones and then migrate into GCs as fully mature GC Tfh cells that serve as persistent virus reservoirs. These findings suggest that viral persistence in lymph nodes drives compensatory differentiation, aberrant accumulation, and latent infection of GC Tfh cells, resulting in marked impairment of humoral immune responses. IMPORTANCE Generation of antibodies that can effectively eliminate viruses requires interactions of B cells with highly specialized T cells in GCs of lymphoid tissues called follicular T helper cells. Here we show that in simian immunodeficiency virus infection, these cells are initially infected in a precursor stage that leads to alterations in their homing, accumulation, and function that may be responsible for the inability of human immunodeficiency virus-infected patients to generate effective antibody responses.
Collapse
|
389
|
Cubas R, van Grevenynghe J, Wills S, Kardava L, Santich BH, Buckner CM, Muir R, Tardif V, Nichols C, Procopio F, He Z, Metcalf T, Ghneim K, Locci M, Ancuta P, Routy JP, Trautmann L, Li Y, McDermott AB, Koup RA, Petrovas C, Migueles SA, Connors M, Tomaras GD, Moir S, Crotty S, Haddad EK. Reversible Reprogramming of Circulating Memory T Follicular Helper Cell Function during Chronic HIV Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:5625-36. [PMID: 26546609 DOI: 10.4049/jimmunol.1501524] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022]
Abstract
Despite the overwhelming benefits of antiretroviral therapy (ART) in curtailing viral load in HIV-infected individuals, ART does not fully restore cellular and humoral immunity. HIV-infected individuals under ART show reduced responses to vaccination and infections and are unable to mount an effective antiviral immune response upon ART cessation. Many factors contribute to these defects, including persistent inflammation, especially in lymphoid tissues, where T follicular helper (Tfh) cells instruct and help B cells launch an effective humoral immune response. In this study we investigated the phenotype and function of circulating memory Tfh cells as a surrogate of Tfh cells in lymph nodes and found significant impairment of this cell population in chronically HIV-infected individuals, leading to reduced B cell responses. We further show that these aberrant memory Tfh cells exhibit an IL-2-responsive gene signature and are more polarized toward a Th1 phenotype. Treatment of functional memory Tfh cells with IL-2 was able to recapitulate the detrimental reprogramming. Importantly, this defect was reversible, as interfering with the IL-2 signaling pathway helped reverse the abnormal differentiation and improved Ab responses. Thus, reversible reprogramming of memory Tfh cells in HIV-infected individuals could be used to enhance Ab responses. Altered microenvironmental conditions in lymphoid tissues leading to altered Tfh cell differentiation could provide one explanation for the poor responsiveness of HIV-infected individuals to new Ags. This explanation has important implications for the development of therapeutic interventions to enhance HIV- and vaccine-mediated Ab responses in patients under ART.
Collapse
Affiliation(s)
- Rafael Cubas
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Julien van Grevenynghe
- Institut National de la Recherche Scientifique, Institut Armand-Frappier, Laval H7V 1B7, Quebec, Canada
| | - Saintedym Wills
- Department of Immunology and the Duke Human Vaccine Institute, Duke University, Durham, NC 27710
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Brian H Santich
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Clarisa M Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Roshell Muir
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Virginie Tardif
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Carmen Nichols
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Francesco Procopio
- Service d'Immunologie et Allergie, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - Zhong He
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Talibah Metcalf
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Khader Ghneim
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Michela Locci
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Petronella Ancuta
- Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, Quebec H3H 2R9, Canada; Research Institute, McGill University Health Centre, Montreal, Quebec H3H 2R9, Canada; Division of Hematology, McGill University Health Centre, Montreal, Quebec H3H 2R9, Canada
| | - Lydie Trautmann
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987
| | - Yuxing Li
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037; Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Adrian B McDermott
- Immunology Laboratory, Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rick A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Constantinos Petrovas
- Immunology Laboratory, Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Steven A Migueles
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mark Connors
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Georgia D Tomaras
- Department of Immunology and the Duke Human Vaccine Institute, Duke University, Durham, NC 27710
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093; and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA 92037
| | - Elias K Haddad
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL 34987;
| |
Collapse
|
390
|
Abstract
BACKGROUND HIV elite controllers suppress HIV viremia without antiretroviral therapy (ART), yet previous studies demonstrated that elite controllers maintain an activated T-cell phenotype. Chronic immune activation has detrimental consequences and thus ART has been advocated for all elite controllers. However, elite controllers are not a clinically homogenous group. Since CD4% is among the best predictors of AIDS-related events, in the current study, we assessed whether this marker can be used to stratify elite controllers needing ART. METHODS Sixteen elite controllers were divided into two groups based on CD4% (EC > 40% and EC ≤40%), and T-cell subsets were analyzed for markers of memory/differentiation (CD45RA, CCR7, CD28), activation (CD38/HLA-DR), immunosenescence (CD57), costimulation (CD73, CD28) and exhaustion (PD-1, CD160, Tim-3). Monocyte subsets (CD14, CD16) were also analyzed and sCD14 levels were quantified using ELISA. RESULTS In the EC group, expression of activation, exhaustion, and immunosensescence markers on T cells were significantly reduced compared with the EC group and similar to the seronegative controls. The EC group expressed higher levels of costimulatory molecules CD28 and CD73 and had lower levels of monocyte activation (HLA-DR expression) with a reduced frequency of inflammatory monocyte (CD14 CD16) subset. Furthermore, the EC group maintained a stable CD4% during a median follow-up of 6 years. CONCLUSION Elite controllers with preserved CD4T cells (EC) have normal T-cell and monocyte phenotypes and therefore may have limited benefit from ART. CD4% can be an important marker for evaluating future studies aimed at determining the need for ART in this group of individuals.
Collapse
|
391
|
Abstract
Chronic viral infections represent a unique challenge to the infected host. Persistently replicating viruses outcompete or subvert the initial antiviral response, allowing the establishment of chronic infections that result in continuous stimulation of both the innate and adaptive immune compartments. This causes a profound reprogramming of the host immune system, including attenuation and persistent low levels of type I interferons, progressive loss (or exhaustion) of CD8(+) T cell functions, and specialization of CD4(+) T cells to produce interleukin-21 and promote antibody-mediated immunity and immune regulation. Epigenetic, transcriptional, posttranscriptional, and metabolic changes underlie this adaptation or recalibration of immune cells to the emerging new environment in order to strike an often imperfect balance between the host and the infectious pathogen. In this review we discuss the common immunological hallmarks observed across a range of different persistently replicating viruses and host species, the underlying molecular mechanisms, and the biological and clinical implications.
Collapse
Affiliation(s)
- Elina I Zuniga
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - Monica Macal
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - Gavin M Lewis
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - James A Harker
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
392
|
Thornhill J, Frater J, Fidler S. Post-treatment control: a functional cure for HIV. Future Virol 2015. [DOI: 10.2217/fvl.15.92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Controlled plasma viral load to levels below limits of detection off antiretroviral therapy (ART), referred to as a functional cure, or remission, is an aspiration for people living with HIV avoiding the need for daily ART while preserving immunological function. For the majority of people, stopping suppressive ART is associated with virological rebound. Spontaneous viral control is reported among rare individuals in the absence of ART. More recently, strategic use of transient ART, initiated close to the time of HIV acquisition, has been associated with a phenotype of viral control after stopping therapy termed post-treatment control. We explore the phenotype of post-treatment viral control, potential underlying mechanisms, and how this area of research can inform HIV cure research.
Collapse
Affiliation(s)
- John Thornhill
- Winston Churchill Wing, St Marys Hospital Imperial College London, Praed St, London, W2 1NY, UK
| | - John Frater
- Winston Churchill Wing, St Marys Hospital Imperial College London, Praed St, London, W2 1NY, UK
- Peter Medwar Building, University of Oxford, S Parks Rd, Oxford OX1 3SY, UK
| | - Sarah Fidler
- Winston Churchill Wing, St Marys Hospital Imperial College London, Praed St, London, W2 1NY, UK
| |
Collapse
|
393
|
Abstract
More than 75 million people worldwide have been infected with human immunodeficiency virus (HIV), and there are now approximately 37 million individuals living with the infection. Untreated HIV replication causes progressive CD4(+) T cell loss and a wide range of immunological abnormalities, leading to an increased risk of infectious and oncological complications. HIV infection also contributes to cardiovascular disease, bone disease, renal and hepatic dysfunction and several other common morbidities. Antiretroviral drugs are highly effective at inhibiting HIV replication, and for individuals who can access and adhere to these drugs, combination antiretroviral therapy leads to durable (and probably lifelong) suppression of viral replication. Viral suppression enables immune recovery and the near elimination of the risk for developing acquired immune deficiency syndrome (AIDS). Despite effective treatment, HIV-infected individuals have a higher than expected risk of heart, bone, liver, kidney and neurological disease. When used optimally by an infected (or by an uninfected) person, antiretroviral drugs can virtually eliminate the risk of HIV transmission. Despite major advances in prevention sciences, HIV transmission remains common in many vulnerable populations, including men who have sex with men, injection drug users and sex workers. Owing to a lack of widespread HIV testing and the costs and toxicities associated with antiretroviral drugs, the majority of the infected population is not on effective antiretroviral therapy. To reverse the pandemic, improved prevention, treatment and implementation approaches are necessary.
Collapse
Affiliation(s)
- Steven G Deeks
- University of California, San Francisco, Department of Medicine, 995 Potrero Avenue, San Francisco, California 94110, USA
| | - Julie Overbaugh
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andrew Phillips
- Department of Infection and Population Health, University College London, London, UK
| | - Susan Buchbinder
- University of California, San Francisco, Department of Medicine, 995 Potrero Avenue, San Francisco, California 94110, USA.,San Francisco Department of Health, San Francisco, California, USA
| |
Collapse
|
394
|
Damouche A, Lazure T, Avettand-Fènoël V, Huot N, Dejucq-Rainsford N, Satie AP, Mélard A, David L, Gommet C, Ghosn J, Noel N, Pourcher G, Martinez V, Benoist S, Béréziat V, Cosma A, Favier B, Vaslin B, Rouzioux C, Capeau J, Müller-Trutwin M, Dereuddre-Bosquet N, Le Grand R, Lambotte O, Bourgeois C. Adipose Tissue Is a Neglected Viral Reservoir and an Inflammatory Site during Chronic HIV and SIV Infection. PLoS Pathog 2015; 11:e1005153. [PMID: 26402858 PMCID: PMC4581628 DOI: 10.1371/journal.ppat.1005153] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 08/14/2015] [Indexed: 12/14/2022] Open
Abstract
Two of the crucial aspects of human immunodeficiency virus (HIV) infection are (i) viral persistence in reservoirs (precluding viral eradication) and (ii) chronic inflammation (directly associated with all-cause morbidities in antiretroviral therapy (ART)-controlled HIV-infected patients). The objective of the present study was to assess the potential involvement of adipose tissue in these two aspects. Adipose tissue is composed of adipocytes and the stromal vascular fraction (SVF); the latter comprises immune cells such as CD4+ T cells and macrophages (both of which are important target cells for HIV). The inflammatory potential of adipose tissue has been extensively described in the context of obesity. During HIV infection, the inflammatory profile of adipose tissue has been revealed by the occurrence of lipodystrophies (primarily related to ART). Data on the impact of HIV on the SVF (especially in individuals not receiving ART) are scarce. We first analyzed the impact of simian immunodeficiency virus (SIV) infection on abdominal subcutaneous and visceral adipose tissues in SIVmac251 infected macaques and found that both adipocytes and adipose tissue immune cells were affected. The adipocyte density was elevated, and adipose tissue immune cells presented enhanced immune activation and/or inflammatory profiles. We detected cell-associated SIV DNA and RNA in the SVF and in sorted CD4+ T cells and macrophages from adipose tissue. We demonstrated that SVF cells (including CD4+ T cells) are infected in ART-controlled HIV-infected patients. Importantly, the production of HIV RNA was detected by in situ hybridization, and after the in vitro reactivation of sorted CD4+ T cells from adipose tissue. We thus identified adipose tissue as a crucial cofactor in both viral persistence and chronic immune activation/inflammation during HIV infection. These observations open up new therapeutic strategies for limiting the size of the viral reservoir and decreasing low-grade chronic inflammation via the modulation of adipose tissue-related pathways.
Collapse
Affiliation(s)
- Abderaouf Damouche
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Thierry Lazure
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’anatomo-pathologie, Le Kremlin-Bicêtre, France
| | - Véronique Avettand-Fènoël
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - Nicolas Huot
- Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | | | - Anne-Pascale Satie
- INSERM, U1085-IRSET, Université de Rennes 1, Campus de Beaulieu, Rennes, France
| | - Adeline Mélard
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - Ludivine David
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | | | - Jade Ghosn
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
| | - Nicolas Noel
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie clinique, Le Kremlin-Bicêtre, France
| | - Guillaume Pourcher
- Assistance Publique—Hôpitaux de Paris, Hôpital Béclère, Service de Chirurgie Viscérale Minimale invasive, Clamart, France
- INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Valérie Martinez
- Assistance Publique—Hôpitaux de Paris, Hôpital Antoine Béclère, Service de Médecine Interne et Immunologie clinique, Clamart, France
| | - Stéphane Benoist
- Assistance Publique—Hôpitaux de Paris, Hôpital Bicêtre, Service de Chirurgie générale et digestive, Le Kremlin-Bicêtre, France
| | - Véronique Béréziat
- INSERM UMR S938, CDR Saint-Antoine; Sorbonne Universités, UPMC Univ Paris 6, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, Service de Biochimie et Hormonologie; ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Antonio Cosma
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Benoit Favier
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Bruno Vaslin
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Christine Rouzioux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - Jacqueline Capeau
- INSERM UMR S938, CDR Saint-Antoine; Sorbonne Universités, UPMC Univ Paris 6, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, Service de Biochimie et Hormonologie; ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | | | - Nathalie Dereuddre-Bosquet
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Olivier Lambotte
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie clinique, Le Kremlin-Bicêtre, France
| | - Christine Bourgeois
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- * E-mail:
| |
Collapse
|
395
|
Chowdhury A, Hayes TL, Bosinger SE, Lawson BO, Vanderford T, Schmitz JE, Paiardini M, Betts M, Chahroudi A, Estes JD, Silvestri G. Differential Impact of In Vivo CD8+ T Lymphocyte Depletion in Controller versus Progressor Simian Immunodeficiency Virus-Infected Macaques. J Virol 2015; 89:8677-86. [PMID: 26063417 PMCID: PMC4524088 DOI: 10.1128/jvi.00869-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED Numerous studies have demonstrated that CD8(+) T lymphocytes suppress virus replication during human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection. However, the mechanisms underlying this activity of T cells remain incompletely understood. Here, we conducted CD8(+) T lymphocyte depletion in 15 rhesus macaques (RMs) infected intravenously (i.v.) with SIVmac239. At day 70 postinfection, the animals (10 progressors with high viremia and 5 controllers with low viremia) were CD8 depleted by i.v. administration of the antibody M-T807R1. As expected, CD8 depletion resulted in increased virus replication, more prominently in controllers than progressors, which correlated inversely with predepletion viremia. Of note, the feature of CD8(+) T lymphocyte predepletion that correlated best with the increase in viremia postdepletion was the level of CD8(+) T-bet(+) lymphocytes. We next found that CD8 depletion resulted in a homogenous increase of SIV RNA in superficial and mesenteric lymph nodes, spleen, and the gastrointestinal tract of both controllers and progressors. Interestingly, the level of SIV DNA increased postdepletion in both CD4(+) central memory T lymphocytes (TCM) and CD4(+) effector memory T lymphocytes (TEM) in progressor RMs but decreased in the CD4(+) TCM of 4 out of 5 controllers. Finally, we found that CD8 depletion is associated with a greater increase in CD4(+) T lymphocyte activation (measured by Ki-67 expression) in controllers than in progressors. Overall, these data reveal a differential impact of CD8(+) T lymphocyte depletion between controller and progressor SIV-infected RMs, emphasizing the complexity of the in vivo antiviral role of CD8(+) T lymphocytes. IMPORTANCE In this study, we further dissect the impact of CD8(+) T lymphocytes on HIV/SIV replication during SIV infection. CD8(+) T lymphocyte depletion leads to a relatively homogenous increase in viral replication in peripheral blood and tissues. CD8(+) T lymphocyte depletion resulted in a more prominent increase in viral loads and CD4(+) T lymphocyte activation in controllers than in progressors. Interestingly, we found T-bet expression on CD8(+) T lymphocytes to be the best predictor of viral load increase following depletion. The levels of SIV DNA increase postdepletion in both CD4(+) TCM and TEM in progressor RMs but decrease in the CD4(+) TCM of controllers. The findings described in this study provide key insights into the differential functions of CD8(+) T lymphocytes in controller and progressor RMs.
Collapse
Affiliation(s)
- Ankita Chowdhury
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Timothy L Hayes
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Steven E Bosinger
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Benton O Lawson
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Thomas Vanderford
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mirko Paiardini
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Michael Betts
- Institute for Immunology and Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann Chahroudi
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
396
|
Glavan TW, Gaulke CA, Hirao LA, Sankaran-Walters S, Dandekar S. SIV-infection-driven changes of pattern recognition receptor expression in mesenteric lymph nodes and gut microbiota dysbiosis. J Med Primatol 2015; 44:241-52. [PMID: 26275157 DOI: 10.1111/jmp.12187] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND The impact of HIV infection on pattern recognition receptor (PRR) expression in gut-associated lymphoid tissue and its association with dysbiosis is not well understood. METHODS PRR and cytokine gene expression were examined in mesenteric lymph nodes (mLN) of rhesus macaques during acute and chronic (untreated and early antiretroviral (ART) treated) infections. Gene expression was correlated with microbial abundance in the gut and immune activation. RESULTS PRR expression rapidly increases during acute infection and is significantly decreased in chronic infection. Early ART maintains elevated PRR expression. Correlation analysis revealed three distinct groups of bacterial taxa that were associated with gene expression changes in infection. CONCLUSIONS PRR and cytokine gene expression in the gut-draining mLN are rapidly modulated in response to viral infection and are correlated with gut dysbiosis. These data suggest that the dysregulation of PRR and related cytokine expression may contribute to chronic immune activation in SIV infection.
Collapse
Affiliation(s)
- Tiffany W Glavan
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | - Christopher A Gaulke
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | - Lauren A Hirao
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | | | - Satya Dandekar
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| |
Collapse
|
397
|
Gadhamsetty S, Beltman JB, de Boer RJ. What do mathematical models tell us about killing rates during HIV-1 infection? Immunol Lett 2015; 168:1-6. [PMID: 26279491 DOI: 10.1016/j.imlet.2015.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/15/2015] [Accepted: 07/31/2015] [Indexed: 10/23/2022]
Abstract
Over the past few decades the extent to which cytotoxic T lymphocytes (CTLs) control human immunodeficiency virus (HIV) replication has been studied extensively, yet their role and mode of action remain controversial. In some studies, CTLs were found to kill a large fraction of the productively infected cells relative to the viral cytopathicity, whereas in others CTLs were suggested to kill only a small fraction of infected cells. In this review, we compile published estimates of CTL-mediated death rates, and examine whether these studies permit determining the rate at which CTLs kill HIV-1 infected cells. We highlight potential misinterpretations of the CTL-killing rates from the escape rates of mutants, and from perturbations of the steady state viral load during chronic infection. Our major conclusion is that CTL-mediated killing rates remain unknown. But contrary to current consensus, we argue that killing rates higher than one per day are perfectly consistent with the experimental data, which would imply that the majority of the productively infected cells could still die from CTL-mediated killing rather than from viral cytopathicity.
Collapse
Affiliation(s)
- Saikrishna Gadhamsetty
- Theoretical Biology and Bioinformatics, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.
| | - Joost B Beltman
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Rob J de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
398
|
Girard MP, Picot V, Longuet C, Nabel GJ. Report of the 2014 Cent Gardes HIV Vaccine Conference-Part 2: Cell-mediated immunity, mucosal protection, and clinical trials: Fondation Mérieux Conference Center, Veyrier du Lac, France, 5-7 October, 2014. Vaccine 2015; 33:4051-5. [PMID: 26143614 DOI: 10.1016/j.vaccine.2015.06.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/04/2015] [Accepted: 06/20/2015] [Indexed: 11/17/2022]
Abstract
The 2014 Cent Gardes Conference took place on October 5-7, 2014, at the Fondation Mérieux Conference Center, on the shores of the Annecy Lake and aimed to review the progress and promise of HIV vaccines. The elicitation of broadly neutralizing antibodies (bNAbs), their use in passive immunization, as well as their genetic delivery (vector immunoprophylaxis) by a recombinant Adenovirus-associated virus (AAV) vector were reviewed in a preceding article [1]. Approaches to the elicitation of long-lasting T cell or mucosal immunity were also discussed and are now reviewed here. The possibility of eliciting mucosal IgAs was discussed, since it was demonstrated that transcytosis-blocking IgAs can protect monkeys against repeated vaginal challenge with a pathogenic chimeric simian and human immunodeficiency virus (SHIV). The possibility of purging the HIV reservoirs from HIV-infected persons and developing a cure of the disease was also discussed.
Collapse
Affiliation(s)
- Marc P Girard
- French National Academy of Medicine, 16 rue Bonaparte, 75006 Paris, France.
| | | | | | - Gary J Nabel
- Sanofi, 640 Memorial Drive, Cambridge, MA 021139, United States.
| |
Collapse
|
399
|
Pinkevych M, Cromer D, Tolstrup M, Grimm AJ, Cooper DA, Lewin SR, Søgaard OS, Rasmussen TA, Kent SJ, Kelleher AD, Davenport MP. HIV Reactivation from Latency after Treatment Interruption Occurs on Average Every 5-8 Days--Implications for HIV Remission. PLoS Pathog 2015; 11:e1005000. [PMID: 26133551 PMCID: PMC4489624 DOI: 10.1371/journal.ppat.1005000] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/03/2015] [Indexed: 12/11/2022] Open
Abstract
HIV infection can be effectively controlled by anti-retroviral therapy (ART) in most patients. However therapy must be continued for life, because interruption of ART leads to rapid recrudescence of infection from long-lived latently infected cells. A number of approaches are currently being developed to 'purge' the reservoir of latently infected cells in order to either eliminate infection completely, or significantly delay the time to viral recrudescence after therapy interruption. A fundamental question in HIV research is how frequently the virus reactivates from latency, and thus how much the reservoir might need to be reduced to produce a prolonged antiretroviral-free HIV remission. Here we provide the first direct estimates of the frequency of viral recrudescence after ART interruption, combining data from four independent cohorts of patients undergoing treatment interruption, comprising 100 patients in total. We estimate that viral replication is initiated on average once every ≈6 days (range 5.1- 7.6 days). This rate is around 24 times lower than previous thought, and is very similar across the cohorts. In addition, we analyse data on the ratios of different 'reactivation founder' viruses in a separate cohort of patients undergoing ART-interruption, and estimate the frequency of successful reactivation to be once every 3.6 days. This suggests that a reduction in the reservoir size of around 50-70-fold would be required to increase the average time-to-recrudescence to about one year, and thus achieve at least a short period of anti-retroviral free HIV remission. Our analyses suggests that time-to-recrudescence studies will need to be large in order to detect modest changes in the reservoir, and that macaque models of SIV latency may have much higher frequencies of viral recrudescence after ART interruption than seen in human HIV infection. Understanding the mean frequency of recrudescence from latency is an important first step in approaches to prolong antiretroviral-free viral remission in HIV.
Collapse
Affiliation(s)
- Mykola Pinkevych
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Deborah Cromer
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Andrew J. Grimm
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - David A. Cooper
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Sharon R. Lewin
- Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Ole S. Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - Miles P. Davenport
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| |
Collapse
|
400
|
Lee SA. Highlights from the Keystone Symposium on HIV Persistence 2015: 26-30 April 2015, Boston, Massachusetts, USA. J Virus Erad 2015; 1:225-6. [PMID: 27482417 PMCID: PMC4946746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|