1
|
Crowell TA. "Let's start at the very beginning": studies of acute HIV inform prevention, diagnosis, and treatment. Curr Opin HIV AIDS 2025; 20:183-185. [PMID: 40178435 DOI: 10.1097/coh.0000000000000930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Affiliation(s)
- Trevor A Crowell
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Parsons MS, Bolton DL. The utility of nonhuman primate models for understanding acute HIV-1 infection. Curr Opin HIV AIDS 2025; 20:218-227. [PMID: 40099824 PMCID: PMC11970610 DOI: 10.1097/coh.0000000000000920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW Nonhuman primate (NHP) models of HIV-1 infection provide complementary experimental pathways for assessing aspects of acute HIV-1 infection (AHI) that cannot be addressed in humans. This article reviews acute infection studies in SIV-infected or SHIV-infected macaque species over the previous 18 months. RECENT FINDINGS Reviewed studies examined the dynamics of replication-competent viral reservoir establishment during early infection, reservoir maintenance throughout therapy, and factors influencing viral rebound after treatment cessation. Also discussed are acute infection events in the central nervous system and liver and potential links between these events and manifestations of comorbidities during chronic infection. Additional studies addressed how occurrences during acute infection impact the development of natural viral control or posttreatment control. Another report evaluated treatment during acute infection with broadly neutralizing antibodies with enhanced ability to engage innate immune cells, highlighting the ability of this early intervention to shape innate and adaptive antiviral responses. SUMMARY NHP models of HIV-1 infection are a fundamental research tool for investigating AHI events. These models enable detailed pathogenesis characterization and the testing of hypothesis-driven strategies for altering disease courses through interventions during AHI, including targeting viral persistence and comorbidities that persist throughout chronic infection.
Collapse
Affiliation(s)
- Matthew S. Parsons
- Walter Reed Army Institute of Research - Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
- U.S. Military HIV Research Program, CIDR, Walter Reed Army Institute of Research, Silver Spring
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Diane L. Bolton
- U.S. Military HIV Research Program, CIDR, Walter Reed Army Institute of Research, Silver Spring
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| |
Collapse
|
3
|
Tiemessen CT. Human models that inform antiretroviral therapy-free remission with perinatally acquired HIV infection. Curr Opin HIV AIDS 2025; 20:249-256. [PMID: 39946194 PMCID: PMC11970615 DOI: 10.1097/coh.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
PURPOSE OF REVIEW Rare persons who achieve disease-control despite high viral loads (viraemic nonprogressors) or maintain virologic control in the absence of antiretroviral therapy (ART) (elite controllers) or following ART interruption (posttreatment controllers) possess protective factors that can be harnessed for interventions to achieve ART-free remission. This review broadly summarizes these phenotypes in adults and children, and updates on findings important in informing strategies for ART-free remission in children with HIV. RECENT FINDINGS To date, only a few individual cases of posttreatment control have been described in children. Smaller HIV reservoir size with very early ART initiation in neonates with in-utero acquired HIV associates with improved virological and immunological outcomes. Nine new cases of ART-free remission in children were recently described - 4 from the P1115 trial, and 5 males from the Ucwaningo Lwabantwana study in South Africa. A striking reduction in the decay of intact proviruses was observed over three decades on suppressive ART in two early-treated twins with HIV. SUMMARY The unique environment of perinatal HIV infection favours effective restriction and decay of the HIV-1 reservoir with suppressive ART initiated very early. Sex and population differences require consideration in ongoing studies to inform ART-free remission.
Collapse
Affiliation(s)
- Caroline T Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
4
|
Essat A, Chapel A, Amokrane K, Monceaux V, Didier C, Melard A, Gardiennet E, Avettand-Fenoel V, Orr S, Boufassa F, Lambotte O, Müller-Trutwin M, Lécuroux C, Chéret A, Goujard C, Rouzioux C, Caillat-Zucman S, Hocqueloux L, Scott-Algara D, Meyer L, Sáez-Cirión A. A genetic fingerprint associated with durable HIV remission after interruption of antiretroviral treatment: ANRS VISCONTI/PRIMO. MED 2025:100670. [PMID: 40300610 DOI: 10.1016/j.medj.2025.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/27/2024] [Accepted: 03/19/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND There is currently no curative treatment for HIV-1 infection. However, some individuals (defined as posttreatment controllers) durably control viremia after the discontinuation of antiretroviral therapy (ART). Although the ability to achieve this HIV-1 remission status is enhanced by early treatment initiation, the mechanisms leading to posttreatment HIV-1 control remain unclear. METHODS We retrospectively explored the immunogenetic characteristics of long-term posttreatment controllers from the ANRS VISCONTI study and persons monitored since primary HIV-1 infection in the ANRS PRIMO cohort and evaluated their influence on clinical parameters and outcome after ART discontinuation. FINDINGS We identified a major histocompatibility complex (MHC)-related fingerprint favoring sustained HIV-1 remission. HLA-B∗35 alleles, which are associated with rapid progression to AIDS during natural HIV-1 infection, were paradoxically overrepresented among posttreatment controllers and had a positive impact on outcome after treatment discontinuation in people who began therapy during primary infection. Specifically, the influence of HLA-B∗35 alleles was observed when they were carried in combination with other HLA class I alleles expressing Bw4 and C2 ligands of killer immunoglobulin-like receptors (KIRs) in a genetic context that favors KIR education of natural killer (NK) cells (Bw4TTC2 genotype). Accordingly, posttreatment controllers with HLA-B∗35 alleles carry distinct KIR genotypes and NK cells. CONCLUSIONS The combination of HLA-B∗35 with Bw4TTC2 genotype, associated with KIR education of NK cells, was abundant among posttreatment HIV-1 controllers and promoted viral control after interruption of early-initiated antiretroviral treatment. These results support a role of NK cells in sustained HIV-1 remission. FUNDING The VISCONTI study and the PRIMO cohort are funded by the ANRS-MIE.
Collapse
Affiliation(s)
- Asma Essat
- Université Paris-Saclay, Inserm CESP U1018, AP-HP, Department of Public Health, Bicêtre Hospital, 94270 Paris-Saclay, France
| | - Anaïs Chapel
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, 75015 Paris, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, 75015 Paris, France
| | - Kahina Amokrane
- AP-HP, Hôpital Saint-Louis, Université Paris Cité, Laboratoire d'Immunologie et Histocompatibilité, 75010 Paris, France
| | - Valérie Monceaux
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, 75015 Paris, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, 75015 Paris, France
| | - Céline Didier
- Institut Pasteur, Unité Régulation des Infections Rétrovirales, 75015 Paris, France
| | - Adeline Melard
- Université Paris Cité, Faculté de Médecine, 75015 Paris, France; INSERM U1016, CNRS UMR8104, Institut Cochin, 75014 Paris, France
| | - Elise Gardiennet
- Université Paris Cité, Faculté de Médecine, 75015 Paris, France; INSERM U1016, CNRS UMR8104, Institut Cochin, 75014 Paris, France
| | - Véronique Avettand-Fenoel
- Université Paris Cité, Faculté de Médecine, 75015 Paris, France; INSERM U1016, CNRS UMR8104, Institut Cochin, 75014 Paris, France; Université d'Orléans, LI(2)RSO, CHU Orléans, Virologie, 45100 Orléans, France
| | - Sylvie Orr
- Université Paris-Saclay, Inserm CESP U1018, AP-HP, Department of Public Health, Bicêtre Hospital, 94270 Paris-Saclay, France
| | - Faroudy Boufassa
- Université Paris-Saclay, Inserm CESP U1018, AP-HP, Department of Public Health, Bicêtre Hospital, 94270 Paris-Saclay, France
| | - Olivier Lambotte
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological, Bacterial Diseases (IMVA-HB/IDMIT/UMRS1184), 94270 Le Kremlin Bicêtre, France
| | - Michaela Müller-Trutwin
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, 75015 Paris, France
| | - Camille Lécuroux
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological, Bacterial Diseases (IMVA-HB/IDMIT/UMRS1184), 94270 Le Kremlin Bicêtre, France
| | - Antoine Chéret
- INSERM U1016, CNRS UMR8104, Institut Cochin, 75014 Paris, France; Université Paris-Saclay, AP-HP, Hôpital Bicêtre, DMU 7, Inserm U1018, CESP, 94270 Le Kremlin Bicêtre, France
| | - Cécile Goujard
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, DMU 7, Inserm U1018, CESP, 94270 Le Kremlin Bicêtre, France
| | | | - Sophie Caillat-Zucman
- AP-HP, Hôpital Saint-Louis, Université Paris Cité, Laboratoire d'Immunologie et Histocompatibilité, 75010 Paris, France
| | - Laurent Hocqueloux
- Centre Hospitalier Universitaire, Service des Maladies Infectieuses, 45100 Orléans, France
| | - Daniel Scott-Algara
- Institut Pasteur, Université Paris Cité, Lymphocyte Cell Biology Unit, 75015 Paris, France
| | - Laurence Meyer
- Université Paris-Saclay, Inserm CESP U1018, AP-HP, Department of Public Health, Bicêtre Hospital, 94270 Paris-Saclay, France
| | - Asier Sáez-Cirión
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, 75015 Paris, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, 75015 Paris, France.
| |
Collapse
|
5
|
Rueger S, Gruener E, Wang D, Abdool FS, Ober V, Vallée T, Stirner R, Conca R, Andrä I, Rogers L, Zahn R, Gersbacher E, Eger J, Pauli R, Postel N, Spinner CD, Vehreschild JJ, Stecher M, Nitschko H, Eberle J, Bogner JR, Seybold U, Draenert R, Leslie A, Kløverpris HN, Geldmacher C, Muenchhoff M, Held K, Roider J. Early treatment and PD1 inhibition enhance HIV-specific functionality of follicular CD8+ T cells. JCI Insight 2025; 10:e180309. [PMID: 40197363 PMCID: PMC11981630 DOI: 10.1172/jci.insight.180309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/18/2025] [Indexed: 04/10/2025] Open
Abstract
People living with HIV treated during acute infection are the group for whom achieving functional cure appears most viable. Follicular CD8+ T cells could contribute to HIV reservoir clearance by accessing B cell follicles through CXCR5 expression. This study examines peripheral follicular CD8+ T cells using flow cytometry, transcriptome analyses, and functional assays in people treated during acute (n = 37) and chronic (n = 18) infection, as well as in individuals naturally controlling HIV (n = 20) and living without HIV (n = 10). Our results reveal that early, as opposed to late, treatment initiation preserves antiviral effector functions of follicular CD8+ T cells, which are further enhanced by PD1 inhibition. We also identify a correlation between follicular CD8+ T cells and intact proviral HIV DNA levels in acute, but not chronic, infection. Longitudinal transcriptomic analysis of peripheral effector cells after 48 weeks of suppressive therapy indicated traits of recent antigen exposure, suggesting potential recirculation into lymphoid tissue. These findings underscore the pivotal role of follicular CD8+ T cells in anti-HIV responses and support investigating targeted cure strategies, such as anti-PD1 therapy, especially in individuals initiating treatment during acute infection.
Collapse
Affiliation(s)
- Susanne Rueger
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Germany
| | - Eva Gruener
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Danni Wang
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Faiaz Shaik Abdool
- Africa Health Research Institute (AHRI), and
- Department of Laboratory Medicine and Medical Science, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Veronica Ober
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Theresa Vallée
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Renate Stirner
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Raffaele Conca
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU University Hospital, LMU Munich, Munich, Germany
| | - Immanuel Andrä
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Lisa Rogers
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Zahn
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Joanna Eger
- Zentrum fuer Innere Medizin und Infektiologie, Munich, Germany
| | | | - Nils Postel
- prinzmed, Practice for Infectiology, Munich, Germany
| | - Christoph D. Spinner
- TUM School of Medicine and Health, Department of Clinical Medicine – Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Jörg J. Vehreschild
- Medical Department 2, Hematology/Oncology and Infectious Diseases, University Hospital of Frankfurt, Frankfurt, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Melanie Stecher
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Hans Nitschko
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Josef Eberle
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Johannes R. Bogner
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Germany
| | - Ulrich Seybold
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rika Draenert
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Antibiotic Stewardship Team, LMU University Hospital, LMU Munich, Munich, Germany
| | - Al Leslie
- Africa Health Research Institute (AHRI), and
- Department of Infection and Immunity, University College London (UCL), London, United Kingdom
| | - Henrik N. Kløverpris
- Africa Health Research Institute (AHRI), and
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Christof Geldmacher
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
| | - Maximilian Muenchhoff
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Kathrin Held
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
- Unit Global Health, Helmholtz Zentrum München, German Research Centre for Environmental Health (HMGU), Neuherberg, Germany
| | - Julia Roider
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Germany
| |
Collapse
|
6
|
Peluso M, Sandel D, Deitchman A, Kim S, Dalhuisen T, Tummala H, Tibúrcio R, Zemelko L, Borgo G, Singh S, Schwartz K, Deswal M, Williams M, Hoh R, Shimoda M, Narpala S, Serebryannyy L, Khalili M, Vendrame E, SenGupta D, Whitmore LS, Tisoncik-Go J, Gale M, Koup R, Mullins J, Felber B, Pavlakis G, Reeves J, Petropoulos C, Glidden D, Spitzer M, Gama L, Caskey M, Nussenzweig M, Chew K, Henrich T, Yukl S, Cohn L, Deeks S, Rutishauser R. Combination immunotherapy induces post-intervention control of HIV. RESEARCH SQUARE 2025:rs.3.rs-6141479. [PMID: 40166020 PMCID: PMC11957202 DOI: 10.21203/rs.3.rs-6141479/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The identification of therapeutic strategies to induce sustained antiretroviral therapy (ART)-free control of HIV infection is a major priority.1 Combination immunotherapy including HIV vaccination, immune stimulation/latency reversal, and passive transfer of broadly neutralizing antibodies (bNAbs) has shown promise in non-human primate models,2-7 but few studies have translated such approaches into people. Here, we performed a single-arm, proof-of-concept combination study of these three approaches in ten people with HIV on ART that included (1) therapeutic vaccination with an HIV/Gag conserved element (CE)-targeted DNA+IL-12 prime/MVA boost regimen followed by (2) administration of two bNAbs (10-1074 and VRC07-523LS) and a toll-like receptor 9 (TLR9) agonist (lefitolimod) during ART suppression, followed by (3) repeat bNAb administration at the time of ART interruption. Seven of the ten participants exhibited partial (low viral load set point) or complete (aviremic) post-intervention control after stopping ART, independent of residual bNAb plasma levels. Robust expansion of activated CD8+ T cells early in response to rebounding virus correlated with lower viral load set points. These data suggest that combination immunotherapy approaches might prove effective to induce sustained control of HIV by slowing rebound and improving CD8+ T cell responses, and that these approaches should continue to be optimized.
Collapse
Affiliation(s)
- M.J Peluso
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - D.A Sandel
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - A.N Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, USA
| | - S.J Kim
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - T Dalhuisen
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - H.P Tummala
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, USA
| | - R Tibúrcio
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - L Zemelko
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - G.M Borgo
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - S.S Singh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - K Schwartz
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - M Deswal
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - M.C Williams
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - R Hoh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - M Shimoda
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - S Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - L Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Khalili
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - E Vendrame
- Gilead Sciences, Inc., Foster City, CA, USA
| | - D SenGupta
- Gilead Sciences, Inc., Foster City, CA, USA
| | - L. S Whitmore
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, USA
| | - J Tisoncik-Go
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, USA
| | - M Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, USA
- Current affiliation: Department of Microbiology and Immunology, and the Institute on Infectious Diseases, University of Minnesota, Minneapolis, MN, USA
| | - R.A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J.I Mullins
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - B.K Felber
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - G.N Pavlakis
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - J.D Reeves
- Labcorp-Monogram Biosciences, South San Francisco, CA, USA
| | | | - D.V Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - M.H Spitzer
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| | - L Gama
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Current affiliation: Instituto Butantan, São Paulo, Brazil
| | - M Caskey
- Department of Clinical Investigation, The Rockefeller University, New York, NY, USA
| | - M.C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - K.W Chew
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - T.J Henrich
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - S.A Yukl
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - L.B Cohn
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - S.G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - R.L Rutishauser
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
7
|
Janssens J, Wedrychowski A, Kim SJ, Isbell C, Hoh R, Pillai SK, Henrich TJ, Deeks SG, Roan NR, Lee SA, Yukl SA. Longitudinal changes in the transcriptionally active and intact HIV reservoir after starting ART during acute infection. J Virol 2025; 99:e0143124. [PMID: 39907283 PMCID: PMC11915860 DOI: 10.1128/jvi.01431-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/25/2024] [Indexed: 02/06/2025] Open
Abstract
Even in antiretroviral therapy (ART)-suppressed human immunodeficiency virus (HIV)-infected individuals, there are heterogeneous populations of HIV-expressing cells exhibiting variable degrees of progression through blocks to HIV transcriptional initiation, elongation, completion, and splicing. These HIV-transcribing cells likely contribute to HIV-associated immune activation and inflammation as well as the viral rebound that occurs after stopping ART. However, it is unclear whether the blocks to HIV transcription are present before ART and how the timing and duration of ART may affect the clearance of cells expressing HIV transcripts that differ in their processivity and/or presence of mutations. To investigate these questions, we quantified different types of HIV transcripts and the corresponding HIV DNA regions/proviruses in longitudinal blood samples obtained before ART initiation (T1) and after 6 months (T2) and 1 year (T3) of ART in 16 individuals who initiated ART during acute HIV infection. Before ART, the pattern of HIV transcripts suggested blocks to elongation and splicing, and only ~10% of intact proviruses were transcribing intact HIV RNA. During the first 6 months of ART, we detected progressively greater reductions in initiated, 5'-elongated, mid-transcribed, completed, and multiply spliced HIV transcripts. Completed HIV RNA decayed faster than initiated or 5'-elongated HIV RNA, and intact HIV RNA tended to decay faster than defective HIV RNA. HIV DNA and RNA levels at T1-T3 correlated inversely with baseline CD4+ T-cell counts. Our findings suggest the existence of immune responses that act selectively to reduce HIV transcriptional completion and/or preferentially kill cells making completed or intact HIV RNA.IMPORTANCEEven in virologically suppressed HIV-infected individuals, expression of viral products from both intact and defective proviruses may contribute to HIV-associated immune activation and inflammation, which are thought to underlie the organ damage that persists despite suppressive ART. We investigated how the timing of ART initiation and the duration of ART affect the heterogeneous populations of HIV-transcribing cells, including a detailed characterization of the different HIV transcripts produced before ART and the rate at which they decay after ART initiation during acute HIV infection. Even during untreated infection, most cells (~90%) have blocks at some stage of transcription. Furthermore, different HIV transcripts decline at different rates on ART, with the fastest decay of cells making completed and intact HIV RNA. Our results suggest that intrinsic or extrinsic immune responses act selectively to either reduce particular stages of HIV transcription or cause selective killing of cells making particular HIV transcripts.
Collapse
Affiliation(s)
- Julie Janssens
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Adam Wedrychowski
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Sun Jin Kim
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Cordelia Isbell
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Satish K. Pillai
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Vitalant Research Institute, San Francisco, California, USA
| | - Timothy J. Henrich
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Nadia R. Roan
- Department of Urology, University of California, San Francisco (UCSF), San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
| | - Sulggi A. Lee
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Steven A. Yukl
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| |
Collapse
|
8
|
Bailón L, Puertas MC, García-Guerrero MC, Moraes-Cardoso I, Aparicio E, Alarcón-Soto Y, Rivero A, Rosen EP, Estes JD, Blanco J, Olvera A, Mothe B, Martinez-Picado J, Moltó J. Impact of Dolutegravir Plus Lamivudine as First-line Antiretroviral Treatment on the Human Immunodeficiency Virus Type 1 Reservoir and Inflammatory Markers in Peripheral Blood. J Infect Dis 2025; 231:600-610. [PMID: 39465671 PMCID: PMC11911789 DOI: 10.1093/infdis/jiae530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND To compare the effects of first-line antiretroviral therapy (ART) with dolutegravir plus lamivudine (DTG + 3TC) versus dolutegravir plus emtricitabine/tenofovir alafenamide (DTG + FTC/TAF) on the human immunodeficiency virus type 1 (HIV-1) reservoir and immune activation biomarkers in people with HIV (PWH). METHODS DUALITY was a 48-week, single-center, randomized, open-label clinical trial in ART-naive PWH, randomized (1:1) to receive ART with DTG + 3TC (2DR group) or DTG + FTC/TAF (3DR group). We measured total and intact proviral HIV-1 DNA, cell-associated RNA in CD4+ T cells, frequency of HIV-infected CD4+ T cells able to produce p24, plasma soluble inflammatory markers, and activation and exhaustion markers in CD4+ and CD8+ T cells. RESULTS Forty-four participants (22 per study arm) were enrolled, with baseline mean (standard deviation) log10 plasma viral load (pVL) 4.4 (0.7) copies/mL and CD4+ T-cell counts of 493 (221) cells/μL. At week 48, all participants had pVL <50 copies/mL at week 48, except for 1 participant in the 2DR group who was resuppressed after treating syphilis. Changes from baseline in reservoir parameters and immune biomarkers were comparable between groups. CONCLUSIONS First-line ART with DTG + 3TC showed similar reductions of HIV-1 persistence parameters and immune markers as DTG + FTC/TAF, supporting DTG/3TC among preferred first-line ART options for PWH.
Collapse
Affiliation(s)
- Lucía Bailón
- Department of Infectious Diseases, Hospital Universitari Germans Trias i Pujol
- Fundació Lluita contra les Infeccions
| | - Maria C Puertas
- Institute for Health Science Research Germans Trias i Pujol, Badalona
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid
- IrsiCaixa, Badalona
| | | | - Igor Moraes-Cardoso
- Institute for Health Science Research Germans Trias i Pujol, Badalona
- IrsiCaixa, Badalona
- Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Cerdanyola del Vallés
| | | | | | - Angel Rivero
- Department of Infectious Diseases, Hospital Universitari Germans Trias i Pujol
- Fundació Lluita contra les Infeccions
- Projecte dels NOMS-Hispanosida, BCN CheckPoint, Barcelona, Spain
| | - Elias P Rosen
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| | - Julià Blanco
- Institute for Health Science Research Germans Trias i Pujol, Badalona
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid
- IrsiCaixa, Badalona
- Department of Infectious Diseases and Immunity, University of Vic–Central University of Catalonia, Vic, Spain
| | - Alex Olvera
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid
- IrsiCaixa, Badalona
- Department of Infectious Diseases and Immunity, University of Vic–Central University of Catalonia, Vic, Spain
- Biosciences Department, Faculty of Sciences and Technology, University of Vic–Central University of Catalonia, Vic, Spain
| | - Beatriz Mothe
- Department of Infectious Diseases, Hospital Universitari Germans Trias i Pujol
- Fundació Lluita contra les Infeccions
- Institute for Health Science Research Germans Trias i Pujol, Badalona
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid
- IrsiCaixa, Badalona
- Department of Infectious Diseases and Immunity, University of Vic–Central University of Catalonia, Vic, Spain
| | - Javier Martinez-Picado
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid
- IrsiCaixa, Badalona
- Department of Infectious Diseases and Immunity, University of Vic–Central University of Catalonia, Vic, Spain
- Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - José Moltó
- Department of Infectious Diseases, Hospital Universitari Germans Trias i Pujol
- Fundació Lluita contra les Infeccions
- Institute for Health Science Research Germans Trias i Pujol, Badalona
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid
| |
Collapse
|
9
|
Reddy K, Lee GQ, Reddy N, Chikowore TJB, Baisley K, Dong KL, Walker BD, Yu XG, Lichterfeld M, Ndung'u T. Differences in HIV-1 reservoir size, landscape characteristics, and decay dynamics in acute and chronic treated HIV-1 Clade C infection. eLife 2025; 13:RP96617. [PMID: 39976231 PMCID: PMC11841988 DOI: 10.7554/elife.96617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
Persisting HIV reservoir viruses in resting CD4 T cells and other cellular subsets are a barrier to cure efforts. Early antiretroviral therapy (ART) enables post-treatment viral control in some cases, but mechanisms remain unclear. We hypothesised that ART initiated before peak viremia impacts HIV-1 subtype C reservoirs. We studied 35 women at high risk of infection from Durban, South Africa, identified with hyperacute HIV by twice-weekly HIV-RNA testing. Participants included 11 starting ART at a median of 456 (297-1203) days post-onset of viremia (DPOV) and 24 at 1 (1-3) DPOV. Peripheral blood mononuclear cells (PBMCs) were used to measured total HIV-1 DNA by droplet digital PCR (ddPCR) and sequence viral reservoir genomes by full-length proviral sequencing (FLIP-seq). ART during hyperacute infection blunted peak viremia (p<0.0001), but contemporaneous total HIV-1 DNA did not differ (p=0.104). Over 1 year, a decline of total HIV-1 DNA was observed in early treated persons (p=0.0004), but not late treated. Among 697 viral genome sequences, the proviral genetic landscape differed between untreated, late treated, and early treated groups. Intact genomes after 1 year were higher in untreated (31%) versus late treated (14%) and early treated (0%). Treatment in both late and early infection caused more rapid decay of intact (13% and 51% per month) versus defective (2% and 35%) viral genomes. However, intact genomes persisted 1 year post chronic treatment but were undetectable with early ART. Early ART also reduced phylogenetic diversity of intact genomes and limited cytotoxic T lymphocyte immune escape variants in the reservoir. Overall, ART initiated in hyperacute HIV-1 subtype C infection did not impact reservoir seeding but was associated with rapid intact viral genome decay, reduced genetic complexity, and limited immune escape, which may accelerate reservoir clearance in combination with other interventional strategies.
Collapse
Affiliation(s)
| | | | - Nicole Reddy
- Africa Health Research InstituteDurbanSouth Africa
- University of KwaZulu-NatalDurbanSouth Africa
| | - Tatenda JB Chikowore
- Africa Health Research InstituteDurbanSouth Africa
- University College LondonLondonUnited Kingdom
| | - Kathy Baisley
- Africa Health Research InstituteDurbanSouth Africa
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Krista L Dong
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-NatalDurbanSouth Africa
- Harvard Medical SchoolBostonUnited States
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-NatalDurbanSouth Africa
- Harvard Medical SchoolBostonUnited States
| | - Xu G Yu
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
- Brigham and Women's HospitalBostonUnited States
| | - Thumbi Ndung'u
- Africa Health Research InstituteDurbanSouth Africa
- University of KwaZulu-NatalDurbanSouth Africa
- University College LondonLondonUnited Kingdom
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-NatalDurbanSouth Africa
| |
Collapse
|
10
|
Magombedze G, Vendrame E, SenGupta D, Geleziunas R, Little S, Smith D, Walker B, Routy JP, Hecht FM, Chun TW, Sneller M, Li JZ, Deeks SG, Peluso MJ. Early Viral Dynamics Predict Human Immunodeficiency Virus Posttreatment Control After Analytic Treatment Interruption. J Infect Dis 2025; 231:e419-e428. [PMID: 39513745 PMCID: PMC11841646 DOI: 10.1093/infdis/jiae551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND A key research priority for developing a human immunodeficiency virus (HIV) cure strategy is to define the viral dynamics and biomarkers associated with sustained posttreatment control. The ability to predict the likelihood of sustained posttreatment control or noncontrol could minimize the time off antiretroviral therapy (ART) for those destined to be controllers and anticipate longer periods off ART for those destined to be controllers. METHODS Mathematical modeling and machine learning were used to characterize virologic predictors of long-term virologic control, using viral kinetics data from several studies in which participants interrupted ART. Predictors of post-ART outcomes were characterized using data accumulated from the time of treatment interruption, replicating real-time data collection in a clinical study, and classifying outcomes as either posttreatment control (plasma viremia, ≤400 copies/mL at 2 of 3 time points for ≥24 weeks) or noncontrol. RESULTS Potential predictors of virologic control were the time to rebound, the rate of initial rebound, and the peak plasma viremia. We found that people destined to be noncontrollers could be identified within 3 weeks of rebound (prediction scores: accuracy, 80%; sensitivity, 82%; specificity, 71%). CONCLUSIONS Given the widespread use of analytic treatment interruption in cure-related trials, these predictors may be useful to increase the safety of analytic treatment interruption through early identification of people who are unlikely to become posttreatment controllers.
Collapse
Affiliation(s)
| | - Elena Vendrame
- Biology Department, Gilead Sciences, Foster City, California, USA
| | - Devi SenGupta
- Biology Department, Gilead Sciences, Foster City, California, USA
| | - Romas Geleziunas
- Biology Department, Gilead Sciences, Foster City, California, USA
| | - Susan Little
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Davey Smith
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Bruce Walker
- The Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Jean-Pierre Routy
- Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre: Glen Site Research Institute, Montreal, Quebec, Canada
| | - Frederick M Hecht
- Osher Center for Integrative Health, University of California, San Francisco, California, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Sneller
- Laboratory of Immunoregulation, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, California, USA
| | - Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
11
|
Laher F, Mahlangu N, Sibiya M. Beliefs about HIV cure: A qualitative study of people living with HIV in Soweto, South Africa. South Afr J HIV Med 2025; 26:1644. [PMID: 39967752 PMCID: PMC11830836 DOI: 10.4102/sajhivmed.v26i1.1644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/22/2024] [Indexed: 02/20/2025] Open
Abstract
Background Rare cases of HIV cure exist. Clinical trials of HIV cure are also underway. However, little is documented about how potential cures are perceived by African people living with HIV, although they are key stakeholders. Objectives We explored knowledge, beliefs, and experiences about HIV cure in Soweto, South Africa. Method We conducted qualitative research with five stratified focus groups (N = 49). Consenting adults living with HIV were eligible. Facilitators asked participants about their knowledge of HIV cure, experience of purported cures, and beliefs about cure possibilities. Transcripts from audio recordings were thematically analysed. Results Participants had knowledge of the concept of cure as eradication, not remission. Three main themes emerged about possible HIV cures. Firstly, hope and scepticism: people feared unequal access to technologies. Secondly, cultural and conventional approaches: there were beliefs in traditional healers, scepticism towards culturally purported cures (e.g. imbiza herbal tonic), and a desire for medical cures to obviate pill burdens. Thirdly, anticipated socio-behavioural effects: beliefs existed that cures might improve happiness, reduce emotional burdens of disclosure, facilitate HIV-free generations, increase risk behaviours, and reduce health checks, but not change societal attitudes to HIV. Conclusion In Soweto, South Africa, people living with HIV hope for medical technologies - such as cure and long-acting treatments - to relieve the biopsychosocial burdens of chronic treatment. Despite treatment knowledge, some people try culturally purported cures for HIV. In HIV cure trials, consent language should avoid 'cure' when remission is meant. Care should address pill burden, and counselling should address sex, substances, exercise, and nutrition.
Collapse
Affiliation(s)
- Fatima Laher
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Naledi Mahlangu
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mbalenhle Sibiya
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
12
|
Gunst JD, Gohil J, Li JZ, Bosch RJ, White Catherine Seamon A, Chun TW, Mothe B, Gittens K, Praiss L, De Scheerder MA, Vandekerckhove L, Escandón K, Thorkelson A, Schacker T, SenGupta D, Brander C, Papasavvas E, Montaner LJ, Martinez-Picado J, Calin R, Castagna A, Muccini C, de Jong W, Leal L, Garcia F, Gruters RA, Tipoe T, Frater J, Søgaard OS, Fidler S. Time to HIV viral rebound and frequency of post-treatment control after analytical interruption of antiretroviral therapy: an individual data-based meta-analysis of 24 prospective studies. Nat Commun 2025; 16:906. [PMID: 39837813 PMCID: PMC11751076 DOI: 10.1038/s41467-025-56116-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
The only current strategy to test efficacy of novel interventions for sustained HIV control without antiretroviral therapy (ART) among people with HIV (PWH) is through an analytical treatment interruption (ATI). Inclusion of 'placebo' controls in ATIs poses ethical, logistical, and economic challenges. To understand viral dynamics and rates of post-treatment control (PTC) after ATI among PWH receiving either placebo or no intervention, we undertook an individual-participant data meta-analysis. In total, 24 eligible prospective studies with 382 individuals with ≥5 plasma HIV RNA viral loads (pVLs) within the first 84 days post-ATI were included. Early-ART was defined as ART initiation within 6 months of HIV acquisition; others were classified as late-ART or unknown. Median age was 42 years, 91% male, 75% white, 45% received early-ART. Median time to pVL >50, >400, and >10,000 copies/mL was 16 days (interquartile range [IQR]:13-25), 21 (IQR:15-28), and 32 (IQR:20-35), respectively. PTC defined as pVL <50 copies/mL at day 84 occurred in 4% (n = 14) of participants (6% early-ART and 1% late-ART). Sustained PTC of pVL <50 copies/ml after 84 days is rare in PWH, especially in those starting ART late. Our findings inform future interventional HIV cure/remission trials on study size and design.
Collapse
Affiliation(s)
- Jesper D Gunst
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Jesal Gohil
- Department of Infectious Disease Imperial College London, Imperial College NIHR BRC, London, UK
| | - Johanthan Z Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronald J Bosch
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Beatriz Mothe
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Hospital Universitari Germans Trias I Pujol, Badalona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Barcelona, Spain
| | - Kathleen Gittens
- Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Praiss
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Linos Vandekerckhove
- HIV Cure Research Center, Department of General Internal Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Kevin Escandón
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, USA
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Ann Thorkelson
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Timothy Schacker
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, USA
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | | - Christian Brander
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Barcelona, Spain
- AELIX Therapeutics, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | | | | | - Javier Martinez-Picado
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Ruxandra Calin
- Department of Infectious Diseases, Tenon Hospital, Sorbonne University, AP-HP, Paris, France
| | - Antonella Castagna
- Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Camilla Muccini
- Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Wesley de Jong
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Lorna Leal
- Infectious Diseases Department, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Felipe Garcia
- Infectious Diseases Department, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Rob A Gruters
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Timothy Tipoe
- Nuffield Dept of Medicine, University of Oxford, UK and NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - John Frater
- Nuffield Dept of Medicine, University of Oxford, UK and NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Ole S Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sarah Fidler
- Department of Infectious Disease Imperial College London, Imperial College NIHR BRC, London, UK
| |
Collapse
|
13
|
Sandel DA, Rutishauser RL, Peluso MJ. Post-intervention control in HIV immunotherapy trials. Curr Opin HIV AIDS 2025; 20:70-79. [PMID: 39494630 PMCID: PMC11620322 DOI: 10.1097/coh.0000000000000890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
PURPOSE OF REVIEW While post-treatment control following interruption of standard-of-care antiretroviral therapy (ART) is well described, post-intervention control following immunotherapy in HIV cure-related clinical trials is less well understood. We provide an overview of recent studies that have identified post-intervention controllers and review the mechanisms that may drive this biologically important phenotype. RECENT FINDINGS Post-intervention controllers have been identified in recent immunotherapy trials testing broadly neutralizing antibodies, immune modulators, modified T cells, checkpoint inhibitors, and gene therapy administered individually or in combination. Currently, there is substantial variability in how each trial defines post-intervention control, as well as in how the mechanisms underlying such control are evaluated. Such mechanisms include ongoing activity of both exogenous and autologous antibodies, as well as changes in HIV-specific T cell function. SUMMARY While no therapeutic strategy to date has succeeded in definitively inducing HIV control, many studies have identified at least a small number of post-intervention controllers. The field would benefit from a standardized approach to defining and reporting this phenotype, as well as standardization in the approach to assessment of how it is achieved. Such efforts would allow for comparisons across clinical trials and could help accelerate efforts toward an HIV cure.
Collapse
Affiliation(s)
| | | | - Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Giron LB, Pasternak AO, Abdel-Mohsen M. Soluble markers of viral rebound and post-treatment HIV control. Curr Opin HIV AIDS 2025; 20:61-69. [PMID: 39392413 PMCID: PMC11620946 DOI: 10.1097/coh.0000000000000889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
PURPOSE OF REVIEW We focus on the different classes of biological molecules measurable in easily accessible bodily fluids that have the potential to serve as biomarkers for the HIV post-treatment controller (PTC) phenotype and/or the timing of viral rebound after stopping antiretroviral therapy (ART). RECENT FINDINGS Various viral components and host factors measurable in body fluids can play crucial roles in understanding and predicting the PTC phenotype. We review recent findings linking viral components, the quantitative and qualitative features of antibodies (including autologous HIV-specific antibodies), markers of inflammation and tissue damage, other host proteins (including hormones such as sex hormones), as well as metabolites, extracellular vesicles, and cell-free DNA to HIV control post-ART interruption. Several of these molecules can or have the potential to predict the time and probability of viral rebound after stopping ART and are biologically active molecules that can directly or indirectly (by modulating immune pressures) impact the size and activity of HIV reservoirs during and post-ART interruption. SUMMARY A comprehensive model combining multiple markers is needed to predict the PTC phenotype. This model can be leveraged to predict and understand the PTC phenotype, which can guide novel curative interventions to replicate this phenotype in post-treatment non-controllers.
Collapse
Affiliation(s)
| | - Alexander O. Pasternak
- Amsterdam UMC, University of Amsterdam, Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, Amsterdam, Netherlands
| | | |
Collapse
|
15
|
Vemparala B, Guedj J, Dixit NM. Advances in the mathematical modeling of posttreatment control of HIV-1. Curr Opin HIV AIDS 2025; 20:92-98. [PMID: 39633541 DOI: 10.1097/coh.0000000000000896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW Several new intervention strategies have shown significant improvements over antiretroviral therapy (ART) in eliciting lasting posttreatment control (PTC) of HIV-1. Advances in mathematical modelling have offered mechanistic insights into PTC and the workings of these interventions. We review these advances. RECENT FINDINGS Broadly neutralizing antibody (bNAb)-based therapies have shown large increases over ART in the frequency and the duration of PTC elicited. Early viral dynamics models of PTC with ART have been advanced to elucidate the underlying mechanisms, including the role of CD8+ T cells. These models characterize PTC as an alternative set-point, with low viral load, and predict routes to achieving it. Large-scale omic datasets have offered new insights into viral and host factors associated with PTC. Correspondingly, new classes of models, including those using learning techniques, have helped exploit these datasets and deduce causal links underlying the associations. Models have also offered insights into therapies that either target the proviral reservoir, modulate immune responses, or both, assessing their translatability. SUMMARY Advances in mathematical modeling have helped better characterize PTC, elucidated and quantified mechanisms with which interventions elicit it, and informed translational efforts.
Collapse
Affiliation(s)
- Bharadwaj Vemparala
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| | | | - Narendra M Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
16
|
Charre C, Merad Y, Avettand-Fenoel V. HIV-1 reservoir landscape of post-treatment control. Curr Opin HIV AIDS 2025; 20:99-108. [PMID: 39484860 DOI: 10.1097/coh.0000000000000891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
PURPOSE OF REVIEW This review explores the viral reservoir landscape in individuals who control viral replication after treatment interruption (TI), designated as post-treatment controllers (PTCs). Identifying their virologic features is crucial to inform drug-free HIV remission strategies. RECENT FINDINGS Traditionally characterized as small, likely due to early treatment, the viral reservoir of PTCs, after TI, exhibits limited transcriptional activity, residual viral replication and subsequent proviral diversity. Intact proviruses are found to be restricted. In nonhuman primate PTCs, this depletion of intact proviruses is already observed in lymph nodes before TI, suggesting that control mechanisms begin during antiretroviral therapy. Furthermore, recent studies suggest immune-driven proviral deep latency associated with repressive epigenetic features and integration sites in PTCs. While molecular mapping of virological features of PTCs is increasingly precise and coupled with in-depth immunologic assays, robust predictive biomarkers of PTCs are still lacking. SUMMARY Despite limited sample sizes and heterogeneous definitions, common virologic features of PTCs include restricted reservoir size and transcriptional activity, fewer intact proviruses and deep proviral latency. Ongoing research using innovative technologies will further elucidate the mechanisms underlying post-treatment control, paving the way for successful HIV cure interventions.
Collapse
Affiliation(s)
- Caroline Charre
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin
- AP-HP, Service de virologie, Hôpital Cochin, Paris
| | - Yanis Merad
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin
- Hospices Civils de Lyon, Service des Maladies Infectieuses et Tropicales, Lyon
| | - Véronique Avettand-Fenoel
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin
- CHU d'Orléans
- Université d'Orléans, LI RSO, Orléans, France
| |
Collapse
|
17
|
Mesquita FS, Li Y, Li JZ. Viral and immune predictors of HIV posttreatment control. Curr Opin HIV AIDS 2025; 20:54-60. [PMID: 39633539 DOI: 10.1097/coh.0000000000000898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW This review focuses on the viral and immune factors influencing HIV posttreatment control (PTC), a rare condition where individuals maintain viral suppression after discontinuing antiretroviral therapy (ART). RECENT FINDINGS Studies demonstrate that early ART initiation leads to smaller HIV reservoirs and delayed viral rebound in PTCs. Virologically, PTCs harbor smaller HIV reservoirs and show lower levels of reservoir transcriptional activity compared with posttreatment noncontrollers. Immunologically, PTCs exhibit distinct T-cell dynamics, with reduced CD4+ and CD8+ T-cell activation and exhaustion, enhanced natural killer (NK) cell activity, and enhanced proliferative responses of HIV-specific CD8+ T cells post-ART interruption. Additionally, humoral immunity, particularly the development of autologous neutralizing antibodies (aNAbs), plays a role in viral control, though broadly neutralizing antibodies (bnAbs) are rare. SUMMARY The mechanisms behind posttreatment control are multifactorial, involving virological and immunological factors. Early ART initiation, a smaller and less transcriptionally active HIV reservoir, and immune responses including proliferative T-cell activity and NK cell function are key contributors to achieving ART-free HIV remission.
Collapse
Affiliation(s)
- Flavio S Mesquita
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yijia Li
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan Z Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Alexandre M, Prague M, Lhomme E, Lelièvre JD, Wittkop L, Richert L, Lévy Y, Thiébaut R. Definition of Virological Endpoints Improving the Design of HIV Cure Strategies Using Analytical Antiretroviral Treatment Interruption. Clin Infect Dis 2024; 79:1447-1457. [PMID: 38819800 DOI: 10.1093/cid/ciae235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Analytical treatment interruption (ATI) is the gold standard in HIV research for assessing the capability of new therapeutic strategies to control viremia without antiretroviral treatment (ART). The viral setpoint is commonly used as endpoint to evaluate their efficacy. However, in line with recommendations from a consensus meeting, to minimize the risk of increased viremia without ART, trials often implement short ATI phases and stringent virological ART restart criteria. This approach can limit the accurate observation of the setpoint. METHODS We analyzed viral dynamics in 235 people with HIV from 3 trials, examining virological criteria during ATI phases. Time-related (eg time to rebound, peak, and setpoint) and viral load magnitude-related criteria (peak, setpoint, and time-averaged AUC [nAUC]) were described. Spearman correlations were analyzed to identify (1) surrogate endpoints for setpoint and (2) optimal virological ART restart criteria mitigating the risks of ART interruption and the evaluation of viral control. RESULTS Comparison of virological criteria between trials showed strong dependencies on ATI design. Similar correlations were found across trials, with nAUC the most strongly correlated with the setpoint, with correlations >0.70. A threshold >100 000 copies/mL for 2 consecutive measures is requested as a virological ART restart criterion. CONCLUSIONS Our results are in line with recommendations and emphasize the benefits of an ATI phase >12 weeks, with regular monitoring, and a virological ART restart criterion of 10 000 copies/mL to limit the risk for patients while capturing enough information to keep nAUC as an optimal proxy to the setpoint.
Collapse
Affiliation(s)
- Marie Alexandre
- Department of Public Health, University of Bordeaux, Inserm U1219 Bordeaux Population Health Research Center, Inria Statistics in Systems Biology and Translational Medicine (SISTM), Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Mélanie Prague
- Department of Public Health, University of Bordeaux, Inserm U1219 Bordeaux Population Health Research Center, Inria Statistics in Systems Biology and Translational Medicine (SISTM), Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Edouard Lhomme
- Department of Public Health, University of Bordeaux, Inserm U1219 Bordeaux Population Health Research Center, Inria Statistics in Systems Biology and Translational Medicine (SISTM), Bordeaux, France
- Vaccine Research Institute, Créteil, France
- Department of Medical information, CHU Bordeaux, Bordeaux, France
| | - Jean-Daniel Lelièvre
- Vaccine Research Institute, Créteil, France
- Inserm U955, Créteil, France
- AP-HP, Hôpital Henri-Mondor Albert-Chenevier, Service d'Immunologie Clinique et Maladies Infectieuses, Créteil, France
| | - Linda Wittkop
- Department of Public Health, University of Bordeaux, Inserm U1219 Bordeaux Population Health Research Center, Inria Statistics in Systems Biology and Translational Medicine (SISTM), Bordeaux, France
- Vaccine Research Institute, Créteil, France
- Department of Medical information, CHU Bordeaux, Bordeaux, France
| | - Laura Richert
- Department of Public Health, University of Bordeaux, Inserm U1219 Bordeaux Population Health Research Center, Inria Statistics in Systems Biology and Translational Medicine (SISTM), Bordeaux, France
- Vaccine Research Institute, Créteil, France
- Department of Medical information, CHU Bordeaux, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Créteil, France
- Inserm U955, Créteil, France
- AP-HP, Hôpital Henri-Mondor Albert-Chenevier, Service d'Immunologie Clinique et Maladies Infectieuses, Créteil, France
| | - Rodolphe Thiébaut
- Department of Public Health, University of Bordeaux, Inserm U1219 Bordeaux Population Health Research Center, Inria Statistics in Systems Biology and Translational Medicine (SISTM), Bordeaux, France
- Vaccine Research Institute, Créteil, France
- Department of Medical information, CHU Bordeaux, Bordeaux, France
| |
Collapse
|
19
|
Gianella S, Yu T, Wang R, Ignacio C, Schanz M, Kouyos RD, Caballero G, Gaitan N, Rawlings S, Kuster H, Metzner KJ, Gandhi RT, Li JZ, Günthard H, Smith DM, Chaillon A. Viral and Immune Risk Factors of HIV Rebound after Interruption of Antiretroviral Therapy. J Infect Dis 2024:jiae585. [PMID: 39661441 DOI: 10.1093/infdis/jiae585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Identifying risk factors for HIV rebound after treatment interruption is crucial for designing effective remission strategies. METHODS Peripheral blood mononuclear cells from participants in the Zurich HIV Primary Infection Cohort (ZPHI, N=73) and ACTG study A5345 (N=44) were analyzed before ART interruption. We measured cell-associated HIV RNA, total HIV DNA, and proviral diversity (env gene). Immune phenotyping was conducted by flow cytometry. Cox proportional hazards (PH) models and penalized Cox PH models with an adaptive LASSO penalty identified risk factors for time to rebound (HIV RNA >1,000 copies/mL). RESULTS Late ART initiation was associated with higher rebound risk (shorter time to rebound), as compared to early ART. Higher pre-ART HIV RNA, total HIV DNA, and increased cellular HIV transcription at the time of ART interruption were associated with higher rebound risk. Higher proviral diversity was associated with higher rebound risk but only among male participants and those enrolled in the ZPHI cohort. Less CD4+ T cells at ART interruption, higher proportions of effector and terminally differentiated T cells, and more activated and exhausted T cells were associated with higher rebound risk, primarily in early treated participants. No significant immunological risk factors were found in participants treated during chronic HIV. In the combined cohort, total HIV DNA and terminally differentiated CD8+ T Cells appeared to be the most relevant risk factors for time to rebound. CONCLUSION These findings underscore the importance of early ART initiation and suggest that tailored interventions based on virologic, immunologic, and demographic factors may help achieve sustained viral suppression.
Collapse
Affiliation(s)
- S Gianella
- Division of Infectious Diseases and Global Public Health University of California San Diego, CA, USA
| | - T Yu
- Harvard Pilgrim Health Care Institution and Harvard Medical School, MA, USA
| | - R Wang
- Harvard Pilgrim Health Care Institution and Harvard Medical School, MA, USA
- Harvard T.H. Chan School of Public Health, MA, USA
| | - C Ignacio
- Division of Infectious Diseases and Global Public Health University of California San Diego, CA, USA
| | - M Schanz
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - R D Kouyos
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Switzerland
| | - G Caballero
- Division of Infectious Diseases and Global Public Health University of California San Diego, CA, USA
| | - N Gaitan
- Division of Infectious Diseases and Global Public Health University of California San Diego, CA, USA
| | | | - H Kuster
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Switzerland
| | - K J Metzner
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Switzerland
| | - Rajesh T Gandhi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Jonathan Z Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - H Günthard
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Switzerland
| | - D M Smith
- Division of Infectious Diseases and Global Public Health University of California San Diego, CA, USA
| | - A Chaillon
- Division of Infectious Diseases and Global Public Health University of California San Diego, CA, USA
| |
Collapse
|
20
|
Daly MB, Dinh C, Holder A, Rudolph D, Ruone S, Swaims-Kohlmeier A, Khalil G, Sharma S, Mitchell J, Condrey J, Kim D, Pan Y, Curtis K, Williams P, Spreen W, Heneine W, García-Lerma JG. SHIV remission in macaques with early treatment initiation and ultra long-lasting antiviral activity. Nat Commun 2024; 15:10550. [PMID: 39632836 PMCID: PMC11618496 DOI: 10.1038/s41467-024-54783-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Studies in SIV-infected macaques show that the virus reservoir is particularly refractory to conventional suppressive antiretroviral therapy (ART). We posit that optimized ART regimens designed to have robust penetration in tissue reservoirs and long-lasting antiviral activity may be advantageous for HIV or SIV remission. Here we treat macaques infected with RT-SHIV with oral emtricitabine/tenofovir alafenamide and long-acting cabotegravir/rilpivirine without (n = 4) or with (n = 4) the immune activator vesatolimod after the initial onset of viremia. We document full suppression in all animals during treatment (4-12 months) and no virus rebound after treatment discontinuation (1.5-2 years of follow up) despite CD8 + T cell depletion. We show efficient multidrug penetration in virus reservoirs and persisting rilpivirine in plasma for 2 years after the last dose. Our results document a type of virus remission that is achieved through early treatment initiation and provision of ultra long-lasting antiviral activity that persists after treatment cessation.
Collapse
Affiliation(s)
- Michele B Daly
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Chuong Dinh
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Angela Holder
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Donna Rudolph
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Susan Ruone
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Alison Swaims-Kohlmeier
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - George Khalil
- Quantitative Sciences and Data Management Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
- Office of Informatics and Data Management, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sunita Sharma
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - James Mitchell
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jillian Condrey
- Comparative Medicine Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Daniel Kim
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Yi Pan
- Quantitative Sciences and Data Management Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Kelly Curtis
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Walid Heneine
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - J Gerardo García-Lerma
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
21
|
Sáez-Cirión A, Mamez AC, Avettand-Fenoel V, Nabergoj M, Passaes C, Thoueille P, Decosterd L, Hentzien M, Perdomo-Celis F, Salgado M, Nijhuis M, Mélard A, Gardiennet E, Lorin V, Monceaux V, Chapel A, Gourvès M, Lechartier M, Mouquet H, Wensing A, Martinez-Picado J, Yerly S, Rougemont M, Calmy A. Sustained HIV remission after allogeneic hematopoietic stem cell transplantation with wild-type CCR5 donor cells. Nat Med 2024; 30:3544-3554. [PMID: 39222660 PMCID: PMC11645271 DOI: 10.1038/s41591-024-03277-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
HIV cure has been reported for five individuals who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) with cells from CCR5Δ32 homozygous donors. By contrast, viral rebound has occurred in other people living with HIV who interrupted antiretroviral treatment after undergoing allo-HSCT, with cells mostly from wild-type CCR5 donors. Here we report the case of a male individual who has achieved durable HIV remission following allo-HSCT with cells from an unrelated HLA-matched (9 of 10 matching for HLA-A, HLA-B, HLA-C, HLA-DRB1 and HLA-DQB1 alleles) wild-type CCR5 donor to treat an extramedullary myeloid tumor. To date, plasma viral load has remained undetectable for 32 months after the interruption of antiretroviral treatment. Treatment with ruxolitinib has been maintained during this period to treat chronic graft-versus-host disease. Low levels of proviral DNA were detected sporadically after allo-HSCT, including defective but not intact HIV DNA. No virus could be amplified in cultures of CD4+ T cells obtained after antiretroviral treatment interruption, while CD4+ T cells remained susceptible to HIV-1 infection in vitro. Declines in HIV antibodies and undetectable HIV-specific T cell responses further corroborate the absence of viral rebound after antiretroviral treatment interruption. These results suggest that HIV remission could be achieved in the context of allo-HSCT with wild-type CCR5.
Collapse
Affiliation(s)
- Asier Sáez-Cirión
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France.
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France.
| | - Anne-Claire Mamez
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Véronique Avettand-Fenoel
- Institut Cochin-CNRS 8104/INSERM U1016/Université de Paris, Paris, France
- LI2RSO, Université d'Orléans, Orléans, France
- Virologie, CHU d'Orléans, Orléans, France
| | | | - Caroline Passaes
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Paul Thoueille
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurent Decosterd
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Maxime Hentzien
- HIV/AIDS Unit, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Federico Perdomo-Celis
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Maria Salgado
- IrsiCaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Monique Nijhuis
- Translational Virology Research Group, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Adeline Mélard
- Institut Cochin-CNRS 8104/INSERM U1016/Université de Paris, Paris, France
| | - Elise Gardiennet
- Institut Cochin-CNRS 8104/INSERM U1016/Université de Paris, Paris, France
| | - Valérie Lorin
- Humoral Immunology Unit, Inserm U1222, Université Paris Cité, Institut Pasteur, Paris, France
| | - Valérie Monceaux
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Anaïs Chapel
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
- HIV Inflammation and Persistence Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Maël Gourvès
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Marine Lechartier
- Viral Reservoirs and Immune Control Unit, Université Paris Cité, Institut Pasteur, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Unit, Inserm U1222, Université Paris Cité, Institut Pasteur, Paris, France
| | - Annemarie Wensing
- Translational Virology Research Group, Department of Global Public Health & Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Javier Martinez-Picado
- IrsiCaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- UVic-UCC, Vic, Spain
- ICREA, Barcelona, Spain
| | - Sabine Yerly
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Alexandra Calmy
- HIV/AIDS Unit, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
22
|
Barbehenn A, Shi L, Shao J, Hoh R, Hartig HM, Pae V, Sarvadhavabhatla S, Donaire S, Sheikhzadeh C, Milush J, Laird GM, Mathias M, Ritter K, Peluso MJ, Martin J, Hecht F, Pilcher C, Cohen SE, Buchbinder S, Havlir D, Gandhi M, Henrich TJ, Hatano H, Wang J, Deeks SG, Lee SA. Rapid biphasic decay of intact and defective HIV DNA reservoir during acute treated HIV disease. Nat Commun 2024; 15:9966. [PMID: 39557853 PMCID: PMC11574060 DOI: 10.1038/s41467-024-54116-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
Despite antiretroviral therapy (ART), HIV persists in latently-infected cells (the HIV reservoir) which decay slowly over time. Here, leveraging >500 longitudinal samples from 67 people living with HIV (PLWH) treated during acute infection, we developed a mathematical model to predict reservoir decay from peripheral CD4 + T cells. Nonlinear generalized additive models demonstrated rapid biphasic decay of intact DNA (week 0-5: t1/2 ~ 2.83 weeks; week 5-24: t1/2 ~ 15.4 weeks) that extended out to 1 year. These estimates were ~5-fold faster than prior decay estimates among chronic treated PLWH. Defective DNA had a similar biphasic pattern, but data were more variable. Predicted intact and defective decay rates were faster for PLWH with earlier timing of ART initiation, higher initial CD4 + T cell count, and lower pre-ART viral load. In this study, we advanced our limited understanding of HIV reservoir decay at the time of ART initiation, informing future curative strategies targeting this critical time.
Collapse
Affiliation(s)
- Alton Barbehenn
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Lei Shi
- Department of Biostatistics, University of California Berkeley, Berkeley, CA, USA
| | - Junzhe Shao
- Department of Biostatistics, University of California Berkeley, Berkeley, CA, USA
| | - Rebecca Hoh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Heather M Hartig
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Vivian Pae
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sannidhi Sarvadhavabhatla
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sophia Donaire
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Caroline Sheikhzadeh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey Milush
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Michael J Peluso
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey Martin
- Department of Biostatistics & Epidemiology, University of California San Francisco, San Francisco, CA, USA
| | - Frederick Hecht
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christopher Pilcher
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Stephanie E Cohen
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Susan Buchbinder
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Diane Havlir
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Monica Gandhi
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Timothy J Henrich
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Hiroyu Hatano
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jingshen Wang
- Department of Biostatistics, University of California Berkeley, Berkeley, CA, USA
| | - Steven G Deeks
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sulggi A Lee
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
23
|
Sun W, Gao C, Gladkov GT, Roseto I, Carrere L, Parsons EM, Gasca-Capote C, Frater J, Fidler S, Yu XG, Lichterfeld M. Footprints of innate immune activity during HIV-1 reservoir cell evolution in early-treated infection. J Exp Med 2024; 221:e20241091. [PMID: 39466203 PMCID: PMC11519379 DOI: 10.1084/jem.20241091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/28/2024] [Accepted: 09/27/2024] [Indexed: 10/29/2024] Open
Abstract
Antiretroviral treatment (ART) initiation during the early stages of HIV-1 infection is associated with a higher probability of maintaining drug-free viral control during subsequent treatment interruptions, for reasons that remain unclear. Using samples from a randomized-controlled human clinical trial evaluating therapeutic HIV-1 vaccines, we here show that early ART commencement is frequently associated with accelerated and efficient selection of genome-intact HIV-1 proviruses in repressive chromatin locations during the first year after treatment initiation. This selection process was unaffected by vaccine-induced HIV-1-specific T cell responses. Single-cell proteogenomic profiling demonstrated that cells harboring intact HIV-1 displayed a discrete phenotypic signature of immune selection by innate immune responses, characterized by a slight but significant upregulation of HLA-C, HLA-G, the IL-10 receptor, and other markers involved in innate immune regulation. Together, these results suggest an accelerated immune selection of viral reservoir cells during early-treated HIV-1 infection that seems at least partially driven by innate immune responses.
Collapse
Affiliation(s)
- Weiwei Sun
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Gregory Takashi Gladkov
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Isabelle Roseto
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Leah Carrere
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth M. Parsons
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Carmen Gasca-Capote
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College and Imperial College NIHR Biomedical Research Centre, London, UK
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
24
|
Hiner CR, Mueller AL, Su H, Goldstein H. Interventions during Early Infection: Opening a Window for an HIV Cure? Viruses 2024; 16:1588. [PMID: 39459922 PMCID: PMC11512236 DOI: 10.3390/v16101588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Although combination antiretroviral therapy (ART) has been a landmark achievement for the treatment of human immunodeficiency virus (HIV), an HIV cure has remained elusive. Elimination of latent HIV reservoirs that persist throughout HIV infection is the most challenging barrier to an HIV cure. The progressive HIV infection is marked by the increasing size and diversity of latent HIV reservoirs until an effective immune response is mobilized, which can control but not eliminate HIV infection. The stalemate between HIV replication and the immune response is manifested by the establishment of a viral set point. ART initiation during the early stage limits HIV reservoir development, preserves immune function, improves the quality of life, and may lead to ART-free viral remission in a few people living with HIV (PLWH). However, for the overwhelming majority of PLWH, early ART initiation alone does not cure HIV, and lifelong ART is needed to sustain viral suppression. A critical area of research is focused on determining whether HIV could be functionally cured if additional treatments are provided alongside early ART. Several HIV interventions including Block and Lock, Shock and Kill, broadly neutralizing antibody (bNAb) therapy, adoptive CD8+ T cell therapy, and gene therapy have demonstrated delayed viral rebound and/or viral remission in animal models and/or some PLWH. Whether or not their application during early infection can improve the success of HIV remission is less studied. Herein, we review the current state of clinical and investigative HIV interventions and discuss their potential to improve the likelihood of post-treatment remission if initiated during early infection.
Collapse
Affiliation(s)
- Christopher R. Hiner
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - April L. Mueller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - Hang Su
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - Harris Goldstein
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
25
|
Reddy K, Lee GQ, Reddy N, Chikowore TJ, Baisley K, Dong KL, Walker BD, Yu XG, Lichterfeld M, Ndung’u T. Differences in HIV-1 reservoir size, landscape characteristics and decay dynamics in acute and chronic treated HIV-1 Clade C infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.16.24302713. [PMID: 38947072 PMCID: PMC11213047 DOI: 10.1101/2024.02.16.24302713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Persisting HIV reservoir viruses in resting CD4 T cells and other cellular subsets are the main barrier to cure efforts. Antiretroviral therapy (ART) intensification by early initiation has been shown to enable post-treatment viral control in some cases but the underlying mechanisms are not fully understood. We hypothesized that ART initiated during the hyperacute phase of infection before peak will affect the size, decay dynamics and landscape characteristics of HIV-1 subtype C viral reservoirs. Methods We studied 35 women at high risk of infection from Durban, South Africa identified with hyperacute HIV infection by twice weekly testing for plasma HIV-1 RNA. Study participants included 11 who started ART at a median of 456 (297-1203) days post onset of viremia (DPOV), and 24 who started ART at a median of 1 (1-3) DPOV. We used peripheral blood mononuclear cells (PBMC) to measure total HIV-1 DNA by ddPCR and to sequence reservoir viral genomes by full length individual proviral sequencing (FLIP-seq) from onset of detection of HIV up to 1 year post treatment initiation. Results Whereas ART in hyperacute infection blunted peak viremia compared to untreated individuals (p<0.0001), there was no difference in total HIV-1 DNA measured contemporaneously (p=0.104). There was a steady decline of total HIV DNA in early treated persons over 1 year of ART (p=0.0004), with no significant change observed in the late treated group. Total HIV-1 DNA after one year of treatment was lower in the early treated compared to the late treated group (p=0.02). Generation of 697 single viral genome sequences revealed a difference in the longitudinal proviral genetic landscape over one year between untreated, late treated, and early treated infection: the relative contribution of intact genomes to the total pool of HIV-1 DNA after 1 year was higher in untreated infection (31%) compared to late treated (14%) and early treated infection (0%). Treatment initiated in both late and early infection resulted in a more rapid decay of intact (13% and 51% per month) versus defective (2% and 35% per month) viral genomes. However, intact genomes were still observed one year post chronic treatment initiation in contrast to early treatment where intact genomes were no longer detectable. Moreover, early ART reduced phylogenetic diversity of intact genomes and limited the seeding and persistence of cytotoxic T lymphocyte immune escape variants in the reservoir. Conclusions Overall, our results show that whereas ART initiated in hyperacute HIV-1 subtype C infection did not impact reservoir seeding, it was nevertheless associated with more rapid decay of intact viral genomes, decreased genetic complexity and immune escape in reservoirs, which could accelerate reservoir clearance when combined with other interventional strategies.
Collapse
Affiliation(s)
- Kavidha Reddy
- Africa Health Research Institute, Durban, South Africa
| | | | - Nicole Reddy
- Africa Health Research Institute, Durban, South Africa
- University of KwaZulu-Natal, Durban, South Africa
| | - Tatenda J.B. Chikowore
- Africa Health Research Institute, Durban, South Africa
- University College of London, London, UK
| | - Kathy Baisley
- Africa Health Research Institute, Durban, South Africa
- London School of Hygiene and Tropical Medicine, London, UK
| | - Krista L. Dong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Harvard Medical School, Boston, Massachusetts, USA
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | - Thumbi Ndung’u
- Africa Health Research Institute, Durban, South Africa
- University of KwaZulu-Natal, Durban, South Africa
- University College of London, London, UK
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- HIV Pathogenesis Programme (HPP), The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
26
|
Bengu N, Cromhout G, Adland E, Govender K, Herbert N, Lim N, Fillis R, Sprenger K, Vieira V, Kannie S, van Lobenstein J, Chinniah K, Kapongo C, Bhoola R, Krishna M, Mchunu N, Pascucci GR, Cotugno N, Palma P, Tagarro A, Rojo P, Roider J, Garcia-Guerrero MC, Ochsenbauer C, Groll A, Reddy K, Giaquinto C, Rossi P, Hong S, Dong K, Ansari MA, Puertas MC, Ndung'u T, Capparelli E, Lichterfeld M, Martinez-Picado J, Kappes JC, Archary M, Goulder P. Sustained aviremia despite anti-retroviral therapy non-adherence in male children after in utero HIV transmission. Nat Med 2024; 30:2796-2804. [PMID: 38843818 PMCID: PMC11485204 DOI: 10.1038/s41591-024-03105-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
After sporadic reports of post-treatment control of HIV in children who initiated combination anti-retroviral therapy (cART) early, we prospectively studied 284 very-early-cART-treated children from KwaZulu-Natal, South Africa, after vertical HIV transmission to assess control of viremia. Eighty-four percent of the children achieved aviremia on cART, but aviremia persisting to 36 or more months was observed in only 32%. We observed that male infants have lower baseline plasma viral loads (P = 0.01). Unexpectedly, a subset (n = 5) of males maintained aviremia despite unscheduled complete discontinuation of cART lasting 3-10 months (n = 4) or intermittent cART adherence during 17-month loss to follow-up (n = 1). We further observed, in vertically transmitted viruses, a negative correlation between type I interferon (IFN-I) resistance and viral replication capacity (VRC) (P < 0.0001) that was markedly stronger for males than for females (r = -0.51 versus r = -0.07 for IFN-α). Although viruses transmitted to male fetuses were more IFN-I sensitive and of higher VRC than those transmitted to females in the full cohort (P < 0.0001 and P = 0.0003, respectively), the viruses transmitted to the five males maintaining cART-free aviremia had significantly lower replication capacity (P < 0.0001). These data suggest that viremic control can occur in some infants with in utero-acquired HIV infection after early cART initiation and may be associated with innate immune sex differences.
Collapse
Affiliation(s)
- Nomonde Bengu
- Queen Nandi Regional Hospital, Empangeni, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Gabriela Cromhout
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Paediatrics, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Nicholas Lim
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Rowena Fillis
- Harry Gwala Regional Hospital, Pietermaritzburg, South Africa
| | - Kenneth Sprenger
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Samantha Kannie
- General Justice Gizenga Mpanza Regional Hospital, Stanger, South Africa
| | | | | | | | - Roopesh Bhoola
- Harry Gwala Regional Hospital, Pietermaritzburg, South Africa
| | - Malini Krishna
- Harry Gwala Regional Hospital, Pietermaritzburg, South Africa
| | - Noxolo Mchunu
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Giuseppe Rubens Pascucci
- Clinical Immunology and Vaccinology Unit, IRCCS, Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Probiomics S.r.l., Rome, Italy
| | - Nicola Cotugno
- Clinical Immunology and Vaccinology Unit, IRCCS, Ospedale Pediatrico Bambino Gesù, Rome, Italy
- University of Rome Tor Vergata, Rome, Italy
| | - Paolo Palma
- Clinical Immunology and Vaccinology Unit, IRCCS, Ospedale Pediatrico Bambino Gesù, Rome, Italy
- University of Rome Tor Vergata, Rome, Italy
| | - Alfredo Tagarro
- Fundación de Investigación Biomédica Hospital 12 de Octubre, Instituto de Investigación 12 de Octubre (imas12), Madrid, Spain
- Department of Pediatrics, Infanta Sofia University Hospital and Henares University Hospital Foundation for Biomedical Research and Innovation, Madrid, Spain
- Universidad Europea de Madrid, Madrid, Spain
| | - Pablo Rojo
- Fundación de Investigación Biomédica Hospital 12 de Octubre, Instituto de Investigación 12 de Octubre (imas12), Madrid, Spain
| | | | | | | | | | - Kavidha Reddy
- Africa Health Research Institute, Durban, South Africa
| | | | - Paolo Rossi
- Clinical Immunology and Vaccinology Unit, IRCCS, Ospedale Pediatrico Bambino Gesù, Rome, Italy
- University of Rome Tor Vergata, Rome, Italy
| | - Seohyun Hong
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
| | - Krista Dong
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
| | - M Azim Ansari
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Maria C Puertas
- IrsiCaixa AIDS Research Institute, Barcelona, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute, Durban, South Africa
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
- Division of Infection and Immunity, University College London, London, UK
| | | | | | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Barcelona, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Infectious Diseases and Immunity Department, University of Vic-Central University of Catalonia, Vic, Spain
| | - John C Kappes
- University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham Veterans Affairs Medical Center, Research Service, Birmingham, AL, USA
| | - Moherndran Archary
- Department of Paediatrics, University of KwaZulu-Natal, Durban, South Africa
| | - Philip Goulder
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- Africa Health Research Institute, Durban, South Africa.
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA.
| |
Collapse
|
27
|
Kuhn L, Barnabas S, Cotugno N, Peay H, Goulder P, Cotton M, Violari A, Pahwa S, Reddy K, Tagarro A, Otwombe K, Fry S, Vaz P, Lain MG, Nhampossa T, Archary M, Maiga AI, Puthanakit T, Kityo CM, Foster C, Rojo P, Klein N, Nastouli E, Tiemessen CT, de Rossi A, Ndung'u T, Persaud D, Lichterfeld M, Giaquinto C, Palma P, Rossi P. Analytical treatment interruption in children living with HIV: position statement from the EPIICAL consortium. Lancet HIV 2024; 11:e700-e710. [PMID: 39059402 DOI: 10.1016/s2352-3018(24)00157-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/16/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024]
Abstract
Analytical treatment interruption (ATI) is widely acknowledged as an essential component of studies to advance our understanding of HIV cure, but discussion has largely been focused on adults. To address this gap, we reviewed evidence related to the safety and utility of ATI in paediatric populations. Three randomised ATI trials using CD4 T-cell and clinical criteria to guide restart of antiretroviral therapy (ART) have been conducted. These trials found low risks associated with ATI in children, including reassuring findings pertaining to neurocognitive outcomes. Similar to adults treated during acute infection, infants treated early in life have shifts in virological and immunological parameters that increase their likelihood of achieving ART-free viral control. Early ART limits the size and diversity of the viral reservoir and shapes effective innate and HIV-specific humoral and cellular responses. Several cases of durable ART-free viral control in early treated children have been reported. We recommend that, where appropriate for the study question and where adequate monitoring is available, ATI should be integrated into ART-free viral control research in children living with HIV. Paediatric participants have the greatest likelihood of benefiting and potentially the most years to prospectively realise those benefits. Excluding children from ATI trials limits the evidence base and delays access to interventions.
Collapse
Affiliation(s)
- Louise Kuhn
- Gertrude H Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA.
| | - Shaun Barnabas
- Family Centre for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg Academic Hospital, Cape Town, South Africa
| | - Nicola Cotugno
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | | | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, UK; HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa; Africa Health Research Institute, Durban, South Africa; Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
| | - Mark Cotton
- Family Centre for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg Academic Hospital, Cape Town, South Africa
| | - Avy Violari
- Perinatal HIV Research Unit, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Soweto, South Africa; School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kavidha Reddy
- Africa Health Research Institute, Durban, South Africa
| | - Alfredo Tagarro
- Fundación de Investigación Biomédica, Hospital 12 de Octubre, Instituto de Investigación 12 de Octubre, Madrid, Spain; Department of Pediatrics, Infanta Sofía University Hospital, Fundación para la Investigación Biomédica e Innovación Hospital Universitario Infanta Sofía y Hospital del Henares, Madrid, Spain; Universidad Europea de Madrid, Madrid, Spain
| | - Kennedy Otwombe
- Perinatal HIV Research Unit, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Soweto, South Africa; School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Samantha Fry
- Family Centre for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg Academic Hospital, Cape Town, South Africa
| | - Paula Vaz
- Fundação Ariel Glaser contra o SIDA Pediátrico, Maputo, Mozambique
| | | | | | - Moherndran Archary
- Africa Health Research Institute, Durban, South Africa; Department of Paediatrics and Department of Infectious Diseases, University of KwaZulu Natal, Durban, South Africa
| | - Almoustapha Issiaka Maiga
- Department of Medical Biology, CHU Gabriel Toure, University of Sciences Techniques and Technologies of Bamako, Bamako, Mali
| | - Thanyawee Puthanakit
- Department of Pediatrics and Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Caroline Foster
- Department of Paediatric Infectious Diseases, Imperial College Healthcare NHS Trust, London, UK
| | - Pablo Rojo
- Universidad Complutense Madrid, Hospital 12 de Octubre, Instituto de Investigación 12 de Octubre, Madrid, Spain
| | - Nigel Klein
- Africa Health Research Institute, Durban, South Africa; Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Eleni Nastouli
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Caroline T Tiemessen
- Centre for HIV and STIs, National Institutes of Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Anita de Rossi
- Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padua, Padua, Italy
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa; Africa Health Research Institute, Durban, South Africa; Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA; Division of Infection and Immunity, University College London, London, UK
| | - Deborah Persaud
- Johns Hopkins University School of Medicine; Department of Pediatrics, Division of Infectious Diseases, Baltimore, MD, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA; Infectious Disease Division, Brigham and Women's Hospital Harvard, Cambridge, MA, USA
| | - Carlo Giaquinto
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Paolo Rossi
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| |
Collapse
|
28
|
Mihealsick E, Word A, Scully EP. The impact of sex on HIV immunopathogenesis and therapeutic interventions. J Clin Invest 2024; 134:e180075. [PMID: 39286972 PMCID: PMC11405047 DOI: 10.1172/jci180075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Globally, the majority of people living with HIV are women or girls, but they have been a minority of participants in clinical trials and observational studies of HIV. Despite this underrepresentation, differences in the pathogenesis of HIV have been observed between men and women, with contributions from both gender- and sex-based factors. These include differences in the risk of HIV acquisition, in viral load set point and immune activation in responses to viremia, and differences in HIV reservoir maintenance. These differences obligate adequate study in both males and females in order to optimize treatments, but also provide a powerful leverage point for delineating the mechanisms of HIV pathogenesis. The shifts in exposure to sex steroid hormones across a lifespan introduce additional complexity, which again can be used to focus on either genetic or hormonal influences as the driver of an outcome. In this Review, we discuss consistent and reproducible differences by sex across the spectrum of HIV, from acquisition through pathogenesis, treatment, and cure, and explore potential mechanisms and gaps in knowledge.
Collapse
Affiliation(s)
| | | | - Eileen P Scully
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Barbehenn A, Shi L, Shao J, Hoh R, Hartig HM, Pae V, Sarvadhavabhatla S, Donaire S, Sheikhzadeh C, Milush J, Laird GM, Mathias M, Ritter K, Peluso MJ, Martin J, Hecht F, Pilcher C, Cohen SE, Buchbinder S, Havlir D, Gandhi M, Henrich TJ, Hatano H, Wang J, Deeks SG, Lee SA. Rapid Biphasic Decay of Intact and Defective HIV DNA Reservoir During Acute Treated HIV Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.27.24304867. [PMID: 38585951 PMCID: PMC10996734 DOI: 10.1101/2024.03.27.24304867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Despite antiretroviral therapy (ART), HIV persists in latently-infected cells ("the reservoir") which decay slowly over time. Here, leveraging >500 longitudinal samples from 67 people with HIV (PWH) treated during acute infection, we developed a novel mathematical model to predict reservoir decay from peripheral CD4+ T cells. Nonlinear generalized additive models demonstrated rapid biphasic decay of intact DNA (week 0-5: t1/2~2.83 weeks; week 5-24: t1/2~15.4 weeks) that extended out to 1 year. These estimates were ~5-fold faster than prior decay estimates among chronic treated PWH. Defective DNA had a similar biphasic pattern, but data were more variable. Predicted intact and defective decay rates were faster for PWH with earlier timing of ART initiation, higher initial CD4+ T cell count, and lower pre-ART viral load. These data add to our limited understanding of HIV reservoir decay at the time of ART initiation, informing future curative strategies targeting this critical time.
Collapse
Affiliation(s)
- Alton Barbehenn
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Lei Shi
- Department of Biostatistics, University of California Berkeley, Berkeley, CA 94110, USA
| | - Junzhe Shao
- Department of Biostatistics, University of California Berkeley, Berkeley, CA 94110, USA
| | - Rebecca Hoh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Heather M. Hartig
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Vivian Pae
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sannidhi Sarvadhavabhatla
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sophia Donaire
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Caroline Sheikhzadeh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jeffrey Milush
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | | | | | | | - Michael J. Peluso
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jeffrey Martin
- Department of Biostatistics & Epidemiology, University of California San Francisco, CA 94158, USA
| | - Frederick Hecht
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Christopher Pilcher
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Stephanie E. Cohen
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
- San Francisco Department of Public Health, San Francisco, CA 94102, USA
| | - Susan Buchbinder
- San Francisco Department of Public Health, San Francisco, CA 94102, USA
| | - Diane Havlir
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Monica Gandhi
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Timothy J. Henrich
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Hiroyu Hatano
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jingshen Wang
- Department of Biostatistics, University of California Berkeley, Berkeley, CA 94110, USA
| | - Steven G. Deeks
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sulggi A. Lee
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
30
|
Vemparala B, Chowdhury S, Guedj J, Dixit NM. Modelling HIV-1 control and remission. NPJ Syst Biol Appl 2024; 10:84. [PMID: 39117718 PMCID: PMC11310323 DOI: 10.1038/s41540-024-00407-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Remarkable advances are being made in developing interventions for eliciting long-term remission of HIV-1 infection. The success of these interventions will obviate the need for lifelong antiretroviral therapy, the current standard-of-care, and benefit the millions living today with HIV-1. Mathematical modelling has made significant contributions to these efforts. It has helped elucidate the possible mechanistic origins of natural and post-treatment control, deduced potential pathways of the loss of such control, quantified the effects of interventions, and developed frameworks for their rational optimization. Yet, several important questions remain, posing challenges to the translation of these promising interventions. Here, we survey the recent advances in the mathematical modelling of HIV-1 control and remission, highlight their contributions, and discuss potential avenues for future developments.
Collapse
Affiliation(s)
- Bharadwaj Vemparala
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| | - Shreya Chowdhury
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| | - Jérémie Guedj
- Université Paris Cité, IAME, INSERM, F-75018, Paris, France
| | - Narendra M Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India.
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
31
|
Su H, Mueller A, Goldstein H. Recent advances on anti-HIV chimeric antigen receptor-T-cell treatment to provide sustained HIV remission. Curr Opin HIV AIDS 2024; 19:169-178. [PMID: 38695148 PMCID: PMC11981014 DOI: 10.1097/coh.0000000000000858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW Successful sustained remission of HIV infection has been achieved after CCR5Δ32/Δ32 allogeneic hematopoietic stem cell transplantation for treatment of leukemia in a small cohort of people living with HIV (PLWH). This breakthrough demonstrated that the goal of curing HIV was achievable. However, the high morbidity and mortality associated with bone marrow transplantation limits the routine application of this approach and provides a strong rationale for pursuing alternative strategies for sustained long-term antiretroviral therapy (ART)-free HIV remission. Notably, long-term immune-mediated control of HIV replication observed in elite controllers and posttreatment controllers suggests that potent HIV-specific immune responses could provide sustained ART-free remission in PLWH. The capacity of chimeric antigen receptor (CAR)-T cells engineered to target malignant cells to induce remission and cure in cancer patients made this an attractive approach to provide PLWH with a potent HIV-specific immune response. Here, we review the recent advances in the design and application of anti-HIV CAR-T-cell therapy to provide a functional HIV cure. RECENT FINDINGS HIV reservoirs are established days after infection and persist through clonal expansion of infected cells. The continuous interaction between latently infected cells and the immune system shapes the landscape of HIV latency and likely contributes to ART-free viral control in elite controllers. CAR-T cells can exhibit superior antiviral activity as compared with native HIV-specific T cells, particularly because they can be engineered to have multiple HIV specificities, resistance to HIV infection, dual costimulatory signaling, immune checkpoint inhibitors, stem cell derivation, CMV TCR coexpression, and tissue homing ligands. These modifications can significantly improve the capacities of anti-HIV CAR-T cells to prevent viral escape, resist HIV infection, and enhance cytotoxicity, persistence, and tissue penetration. Collectively, these novel modifications of anti-HIV CAR-T cell design have increased their capacity to control HIV infection. SUMMARY Anti-HIV CAR-T cells can be engineered to provide potent and sustained in-vitro and in-vivo antiviral function. The combination of anti-HIV CAR-T cells with other immunotherapeutics may contribute to long-term HIV remission in PLWH.
Collapse
Affiliation(s)
- Hang Su
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
| | - April Mueller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
| | - Harris Goldstein
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, 10461, U.S.A
| |
Collapse
|
32
|
Phan T, Conway JM, Pagane N, Kreig J, Sambaturu N, Iyaniwura S, Li JZ, Ribeiro RM, Ke R, Perelson AS. Understanding early HIV-1 rebound dynamics following antiretroviral therapy interruption: The importance of effector cell expansion. PLoS Pathog 2024; 20:e1012236. [PMID: 39074163 PMCID: PMC11309407 DOI: 10.1371/journal.ppat.1012236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/08/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Most people living with HIV-1 experience rapid viral rebound once antiretroviral therapy is interrupted; however, a small fraction remain in viral remission for an extended duration. Understanding the factors that determine whether viral rebound is likely after treatment interruption can enable the development of optimal treatment regimens and therapeutic interventions to potentially achieve a functional cure for HIV-1. We built upon the theoretical framework proposed by Conway and Perelson to construct dynamic models of virus-immune interactions to study factors that influence viral rebound dynamics. We evaluated these models using viral load data from 24 individuals following antiretroviral therapy interruption. The best-performing model accurately captures the heterogeneity of viral dynamics and highlights the importance of the effector cell expansion rate. Our results show that post-treatment controllers and non-controllers can be distinguished based on the effector cell expansion rate in our models. Furthermore, these results demonstrate the potential of using dynamic models incorporating an effector cell response to understand early viral rebound dynamics post-antiretroviral therapy interruption.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Jessica M. Conway
- Department of Mathematics, Pennsylvania State University, College Township, Pennsylvania, United States of America
- Department of Biology, Pennsylvania State University, College Township, Pennsylvania, United States of America
| | - Nicole Pagane
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, Massachusetts, United States of America
| | - Jasmine Kreig
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Narmada Sambaturu
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Sarafa Iyaniwura
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Ruian Ke
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| |
Collapse
|
33
|
Sugiyama FHC, Dietz LL, Søgaard OS. Utilizing immunotherapy towards achieving a functional cure for HIV-1. Curr Opin HIV AIDS 2024; 19:187-193. [PMID: 38686856 DOI: 10.1097/coh.0000000000000856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
PURPOSE OF REVIEW Advancements in antiretroviral therapy (ART) have positively impacted the life expectancy and possibility of living a normal life for people with HIV-1. However, lifelong daily medication is necessary to prevent disease progression. To this end, immunotherapeutic strategies are being tested with the aim of developing a functional cure in which the immune system effectively controls HIV-1 in the absence of ART. RECENT FINDINGS The most promising advances in achieving sustained HIV-1 remission or cure include broadly neutralizing antibodies (bNAbs) that are administered alone or in combination with other agents. Newer and more innovative approaches redirecting T cells or natural killer cells to kill HIV-1 infected cells have also shown promising results. Finally, multiple ongoing trials focus on combining bNAbs with other immune-directed therapies to enhance both innate and adaptive immunity. SUMMARY While immunotherapies as an alternative to conventional ART have generally proven to be well tolerated, these therapeutic approaches have largely been unsuccessful in inducing ART-free control of HIV-1. However, promising results from recent trials involving bNAbs that have reported durable HIV-1 control among a subset of participants, provide reason for cautious optimism that we with further optimization of these treatment strategies may be able to achieve functional cure for HIV-1.
Collapse
Affiliation(s)
- Fabrícia Heloisa Cavicchioli Sugiyama
- Department of Clinical, Toxicological and Bromatological Analysis, University of São Paulo, Ribeirão Preto, Brazil
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lisa Loksø Dietz
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ole Schmeltz Søgaard
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
34
|
Phan T, Conway JM, Pagane N, Kreig J, Sambaturu N, Iyaniwura S, Li JZ, Ribeiro RM, Ke R, Perelson AS. Understanding early HIV-1 rebound dynamics following antiretroviral therapy interruption: The importance of effector cell expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592318. [PMID: 38746144 PMCID: PMC11092759 DOI: 10.1101/2024.05.03.592318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Most people living with HIV-1 experience rapid viral rebound once antiretroviral therapy is interrupted; however, a small fraction remain in viral remission for an extended duration. Understanding the factors that determine whether viral rebound is likely after treatment interruption can enable the development of optimal treatment regimens and therapeutic interventions to potentially achieve a functional cure for HIV-1. We built upon the theoretical framework proposed by Conway and Perelson to construct dynamic models of virus-immune interactions to study factors that influence viral rebound dynamics. We evaluated these models using viral load data from 24 individuals following antiretroviral therapy interruption. The best-performing model accurately captures the heterogeneity of viral dynamics and highlights the importance of the effector cell expansion rate. Our results show that post-treatment controllers and non-controllers can be distinguished based on the effector cell expansion rate in our models. Furthermore, these results demonstrate the potential of using dynamic models incorporating an effector cell response to understand early viral rebound dynamics post-antiretroviral therapy interruption.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Jessica M Conway
- Department of Mathematics, Pennsylvania State University, College Township, PA, USA
- Department of Biology, Pennsylvania State University, College Township, PA, USA
| | - Nicole Pagane
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
| | - Jasmine Kreig
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Narmada Sambaturu
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Sarafa Iyaniwura
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Jonathan Z Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Ruian Ke
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
35
|
van Paassen P, Dijkstra M, Peay HL, Rokx C, Verbon A, Reiss P, Prins JM, Henderson GE, Rennie S, Nieuwkerk PT, de Bree GJ. Perceptions of Rapid Antiretroviral Therapy Initiation Among Participants of The Netherlands Cohort Study on Acute HIV Infection. AIDS Res Hum Retroviruses 2024; 40:286-292. [PMID: 37791419 DOI: 10.1089/aid.2022.0169] [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] [Indexed: 10/05/2023] Open
Abstract
Starting antiretroviral therapy (ART) same-day, or as soon as possible after HIV diagnosis is advised in guidelines worldwide. Especially during acute HIV infection (AHI), rapid ART start may be more urgent because of a higher risk of transmission or symptoms of acute retroviral syndrome. During this phase, rapid ART start may have additional benefits for viral reservoir size and host immunity. We explored perceptions of rapid ART start among participants of The Netherlands Cohort Study on Acute HIV infection (NOVA study), who started ART rapidly after diagnosis of AHI. We conducted 20 in-depth qualitative interviews with NOVA study participants between October and December 2018. Data were analyzed thematically, using inductive and iterative coding techniques. Roughly half of the participants stated they felt well-informed about the importance of (rapid) ART. Starting ART rapidly was perceived positively by almost all participants, mostly because of the expected benefits on their health, and to prevent HIV transmission. Rapid ART start was seen as a way to cope with the diagnosis. However, a more negative perception was that rapid ART start confronted participants with their diagnosis, when they were still adjusting to a new situation. Our results show that among people diagnosed during AHI, rapid ART is well-accepted. These results should be encouraging to HIV care providers who encounter people with AHI in their clinical practice and to researchers who carry out cure-related studies, in which early ART is often included. The Clinical Trial Registration number is NCT05728996.
Collapse
Affiliation(s)
- Pien van Paassen
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, Academic Medical Center, Amsterdam, The Netherlands
| | - Maartje Dijkstra
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, Academic Medical Center, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, The Netherlands
| | - Holly L Peay
- RTI International, Research Triangle Park, North Carolina, USA
| | - Casper Rokx
- Department of Internal Medicine and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Annelies Verbon
- Department of Internal Medicine, Division of Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter Reiss
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, Academic Medical Center, Amsterdam, The Netherlands
- Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan M Prins
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, Academic Medical Center, Amsterdam, The Netherlands
| | - Gail E Henderson
- Department of Social Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stuart Rennie
- Department of Social Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
- UNC Bioethics Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Pythia T Nieuwkerk
- Department of Medical Psychology, Amsterdam UMC, Academic Medical Center, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Godelieve J de Bree
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Payra S, Manjhi PK, Singh S, Kumar R, Singh SK, Kumar A, Maharshi V. HIV cure: Are we going to make history? HIV Med 2024; 25:322-331. [PMID: 37821095 DOI: 10.1111/hiv.13557] [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: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND At present, combination antiretroviral therapy (cART) is the mainstay for the treatment of people living with HIV/AIDS. cART can suppress the viral load to a minimal level; however, the possibility of the emergence of full-blown AIDS is always there. In the latter part of the first decade of the 21st century, an HIV-positive person received stem cell transplantation (SCT) for treatment of his haematological malignancy. The patient was able to achieve remission of the haematological condition as well as of HIV following SCT. Thorough investigations of various samples including blood and biopsy could not detect the virus in the person's body. The person was declared to be the first cured case of HIV. LITERATURE SEARCH Over the next decade, a few more similar cases were observed and have recently been declared cured of the infection. A comprehensive search was performed in PubMed, Cochrane library and Google Scholar. Four such additional cases were found in literature. DESCRIPTION & DISCUSSION These cases all share a common proposed mechanism for the HIV cure, that is, transplantation of stem cells from donors carrying a homozygous mutation in a gene encoding for CCR5 (receptor utilized by HIV for entry into the host cell), denoted as CCR5△32. This mutation makes the host immune cells devoid of CCR5, causing the host to acquire resistance against HIV. To the best of our knowledge, this is the first review to look at relevant and updated information of all cured cases of HIV as well as the related landmarks in history and discusses the underlying mechanism(s).
Collapse
Affiliation(s)
- Shuvasree Payra
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Pramod Kumar Manjhi
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Shruti Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Rajesh Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Sunil Kumar Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Alok Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| | - Vikas Maharshi
- Department of Pharmacology, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
37
|
Li JZ, Melberg M, Kittilson A, Abdel-Mohsen M, Li Y, Aga E, Bosch RJ, Wonderlich ER, Kinslow J, Giron LB, Di Germanio C, Pilkinton M, MacLaren L, Keefer M, Fox L, Barr L, Acosta E, Ananworanich J, Coombs R, Mellors J, Deeks S, Gandhi RT, Busch M, Landay A, Macatangay B, Smith DM. Predictors of HIV rebound differ by timing of antiretroviral therapy initiation. JCI Insight 2024; 9:e173864. [PMID: 38329130 PMCID: PMC10967395 DOI: 10.1172/jci.insight.173864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUNDIdentifying factors that predict the timing of HIV rebound after treatment interruption will be crucial for designing and evaluating interventions for HIV remission.METHODSWe performed a broad evaluation of viral and immune factors that predict viral rebound (AIDS Clinical Trials Group A5345). Participants initiated antiretroviral therapy (ART) during chronic (N = 33) or early (N = 12) HIV infection with ≥ 2 years of suppressive ART and restarted ART if they had 2 viral loads ≥ 1,000 copies/mL after treatment interruption.RESULTSCompared with chronic-treated participants, early-treated individuals had smaller and fewer transcriptionally active HIV reservoirs. A higher percentage of HIV Gag-specific CD8+ T cell cytotoxic response was associated with lower intact proviral DNA. Predictors of HIV rebound timing differed between early- versus chronic-treated participants, as the strongest reservoir predictor of time to HIV rebound was level of residual viremia in early-treated participants and intact DNA level in chronic-treated individuals. We also identified distinct sets of pre-treatment interruption viral, immune, and inflammatory markers that differentiated participants who had rapid versus slow rebound.CONCLUSIONThe results provide an in-depth overview of the complex interplay of viral, immunologic, and inflammatory predictors of viral rebound and demonstrate that the timing of ART initiation modifies the features of rapid and slow viral rebound.TRIAL REGISTRATIONClinicalTrials.gov NCT03001128FUNDINGNIH National Institute of Allergy and Infectious Diseases, Merck.
Collapse
Affiliation(s)
- Jonathan Z. Li
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan Melberg
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Autumn Kittilson
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Yijia Li
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Evgenia Aga
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Ronald J. Bosch
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | | | | | - Clara Di Germanio
- University of California, San Francisco, San Francisco, California, USA
- Vitalant Research Institute, San Francisco, California, USA
| | - Mark Pilkinton
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | - Lawrence Fox
- National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Liz Barr
- AIDS Clinical Trials Group Community Scientific Subcommittee, Los Angeles, California, USA
| | | | | | | | - John Mellors
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Steven Deeks
- University of California, San Francisco, San Francisco, California, USA
| | - Rajesh T. Gandhi
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Busch
- Vitalant Research Institute, San Francisco, California, USA
| | - Alan Landay
- Rush University Medical Center, Chicago, Illinois, USA
| | | | - Davey M. Smith
- University of California, San Diego, San Diego, California, USA
| | | |
Collapse
|
38
|
Passaes C, Desjardins D, Chapel A, Monceaux V, Lemaitre J, Mélard A, Perdomo-Celis F, Planchais C, Gourvès M, Dimant N, David A, Dereuddre-Bosquet N, Barrail-Tran A, Gouget H, Guillaume C, Relouzat F, Lambotte O, Guedj J, Müller-Trutwin M, Mouquet H, Rouzioux C, Avettand-Fenoël V, Le Grand R, Sáez-Cirión A. Early antiretroviral therapy favors post-treatment SIV control associated with the expansion of enhanced memory CD8 + T-cells. Nat Commun 2024; 15:178. [PMID: 38212337 PMCID: PMC10784587 DOI: 10.1038/s41467-023-44389-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024] Open
Abstract
HIV remission can be achieved in some people, called post-treatment HIV controllers, after antiretroviral treatment discontinuation. Treatment initiation close to the time of infection was suggested to favor post-treatment control, but the circumstances and mechanisms leading to this outcome remain unclear. Here we evaluate the impact of early (week 4) vs. late (week 24 post-infection) treatment initiation in SIVmac251-infected male cynomolgus macaques receiving 2 years of therapy before analytical treatment interruption. We show that early treatment strongly promotes post-treatment control, which is not related to a lower frequency of infected cells at treatment interruption. Rather, early treatment favors the development of long-term memory CD8+ T cells with enhanced proliferative and SIV suppressive capacity that are able to mediate a robust secondary-like response upon viral rebound. Our model allows us to formally demonstrate a link between treatment initiation during primary infection and the promotion of post-treatment control and provides results that may guide the development of new immunotherapies for HIV remission.
Collapse
Affiliation(s)
- Caroline Passaes
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France.
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France.
| | - Delphine Desjardins
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Anaïs Chapel
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Valérie Monceaux
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Julien Lemaitre
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Adeline Mélard
- Université Paris Cité; INSERM, U1016; CNRS, UMR8104, Paris, France
| | - Federico Perdomo-Celis
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Cyril Planchais
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Maël Gourvès
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France
| | - Nastasia Dimant
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Annie David
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Aurélie Barrail-Tran
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, Service de Pharmacie, Le Kremlin Bicêtre, France
| | - Hélène Gouget
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Céline Guillaume
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Olivier Lambotte
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
- Université Paris-Saclay, AP-HP. Hôpital Bicêtre, Clinical Immunology Department, 94270, Le Kremlin Bicêtre, France
| | - Jérémie Guedj
- Université Paris Cité, IAME, INSERM, F-75018, Paris, France
| | - Michaela Müller-Trutwin
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France
| | - Hugo Mouquet
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Christine Rouzioux
- Université Paris Cité/APHP Hôpital Necker - Enfants Malades, Paris, France
| | - Véronique Avettand-Fenoël
- Université Paris Cité; INSERM, U1016; CNRS, UMR8104, Paris, France
- APHP Hôpital Cochin, Service de Virologie, Paris, France
| | - Roger Le Grand
- Université Paris-Saclay, CEA, INSERM, UMR1184, Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT Department), Fontenay-aux-Roses/Le Kremlin-Bicêtre, France
| | - Asier Sáez-Cirión
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, France.
- Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistence Unit, Paris, France.
| |
Collapse
|
39
|
van Paassen PM, van Pul L, van der Straten K, Buchholtz NV, Grobben M, van Nuenen AC, van Dort KA, Boeser-Nunnink BD, van den Essenburg MD, Burger JA, van Luin M, Jurriaans S, Sanders RW, Swelsen WT, Symons J, Klouwens MJ, Nijhuis M, van Gils MJ, Prins JM, de Bree GJ, Kootstra NA. Virological and immunological correlates of HIV posttreatment control after temporal antiretroviral therapy during acute HIV infection. AIDS 2023; 37:2297-2304. [PMID: 37702421 PMCID: PMC10653294 DOI: 10.1097/qad.0000000000003722] [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: 05/26/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVE People with HIV rarely control viral replication after cessation of antiretroviral therapy (ART). We present a person with HIV with extraordinary posttreatment control (PTC) for over 23 years after temporary ART during acute HIV infection (AHI) leading to a new insight in factors contributing to PTC. DESIGN/METHODS Viral reservoir was determined by HIV qPCR, Intact Proviral DNA Assay, and quantitative viral outgrowth assay. Viral replication kinetics were determined in autologous and donor PBMC. IgG levels directed against HIV envelope and neutralizing antibodies were measured. Immune phenotyping of T cells and HIV-specific T-cell responses were analyzed by flow cytometry. RESULTS The case presented with AHI and a plasma viral load of 2.7 million copies/ml. ART was initiated 2 weeks after diagnosis and interrupted after 26 months. Replicating virus was isolated shortly after start ART. At 18 years after treatment interruption, HIV-DNA in CD4 + T cells and low levels of HIV-RNA in plasma (<5 copies/ml) were detectable. Stable HIV envelope glycoprotein-directed IgG was present during follow-up, but lacked neutralizing activity. Strong antiviral CD8 + T-cell responses, in particular targeting HIV-gag, were detected during 25 years follow-up. Moreover, we found a P255A mutation in an HLA-B∗44 : 02 restricted gag-epitope, which was associated with decreased replication. CONCLUSION We describe an exceptional case of PTC, which is likely associated with sustained potent gag-specific CD8 + T-cell responses in combination with a replication attenuating escape mutation in gag. Understanding the initiation and preservation of the HIV-specific T-cell responses could guide the development of strategies to induce HIV control.
Collapse
Affiliation(s)
- Pien M. van Paassen
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
| | - Lisa van Pul
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
| | - Karlijn van der Straten
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Amsterdam
| | - Ninée V.J.E. Buchholtz
- Department of Medical Microbiology, Translational Virology, University Medical Center Utrecht
| | - Marloes Grobben
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Amsterdam
| | - Ad C. van Nuenen
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
| | - Karel A. van Dort
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
| | - Brigitte D. Boeser-Nunnink
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
| | | | - Judith A. Burger
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Amsterdam
| | - Matthijs van Luin
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht
| | - Suzanne Jurriaans
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Amsterdam
| | - Rogier W. Sanders
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Amsterdam
| | - Wendy T. Swelsen
- Department of Immunogenetics, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Jori Symons
- Department of Medical Microbiology, Translational Virology, University Medical Center Utrecht
| | - Michelle J. Klouwens
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Monique Nijhuis
- Department of Medical Microbiology, Translational Virology, University Medical Center Utrecht
| | - Marit J. van Gils
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Amsterdam
| | - Jan M. Prins
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Godelieve J. de Bree
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Neeltje A. Kootstra
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9
- Amsterdam Institute for Infection and Immunity, Infectious Diseases
| |
Collapse
|
40
|
Lyons DE, Kumar P, Roan NR, Defechereux PA, Feschotte C, Lange UC, Murthy N, Sameshima P, Verdin E, Ake JA, Parsons MS, Nath A, Gianella S, Smith DM, Kallas EG, Villa TJ, Strange R, Mwesigwa B, Furler O’Brien RL, Nixon DF, Ndhlovu LC, Valente ST, Ott M. HIV-1 Remission: Accelerating the Path to Permanent HIV-1 Silencing. Viruses 2023; 15:2171. [PMID: 38005849 PMCID: PMC10674359 DOI: 10.3390/v15112171] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Despite remarkable progress, a cure for HIV-1 infection remains elusive. Rebound competent latent and transcriptionally active reservoir cells persevere despite antiretroviral therapy and rekindle infection due to inefficient proviral silencing. We propose a novel "block-lock-stop" approach, entailing long term durable silencing of viral expression towards an irreversible transcriptionally inactive latent provirus to achieve long term antiretroviral free control of the virus. A graded transformation of remnant HIV-1 in PLWH from persistent into silent to permanently defective proviruses is proposed, emulating and accelerating the natural path that human endogenous retroviruses (HERVs) take over millions of years. This hypothesis was based on research into delineating the mechanisms of HIV-1 latency, lessons from latency reversing agents and advances of Tat inhibitors, as well as expertise in the biology of HERVs. Insights from elite controllers and the availability of advanced genome engineering technologies for the direct excision of remnant virus set the stage for a rapid path to an HIV-1 cure.
Collapse
Affiliation(s)
- Danielle E. Lyons
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Nadia R. Roan
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Urology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Patricia A. Defechereux
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA
| | - Cedric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | | | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA;
- Innovative Genomics Institute, Berkeley, CA 94720, USA
| | - Pauline Sameshima
- Faculty of Education, Lakehead University, Thunder Bay, ON P7B 5E1, Canada;
| | - Eric Verdin
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Julie A. Ake
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA (M.S.P.)
| | - Matthew S. Parsons
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA (M.S.P.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD 20824, USA;
| | - Sara Gianella
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Davey M. Smith
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Esper G. Kallas
- Department of Infectious and Parasitic Diseases, University of Sao Paulo, São Paulo 04023-900, Brazil
| | - Thomas J. Villa
- HOPE Martin Delaney Collaboratory for HIV Cure Research Community Engagement Ambassador, Washinton, DC 20004, USA (R.S.)
- National HIV & Aging Advocacy Network, Washington, DC 20004, USA
| | - Richard Strange
- HOPE Martin Delaney Collaboratory for HIV Cure Research Community Engagement Ambassador, Washinton, DC 20004, USA (R.S.)
| | - Betty Mwesigwa
- Research Department, Makerere University Walter Reed Project, Kampala P.O Box 7062, Uganda
| | - Robert L. Furler O’Brien
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Douglas F. Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Susana T. Valente
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL 33458, USA
| | - Melanie Ott
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
41
|
Kopycinski J, Yang H, Hancock G, Pace M, Kim E, Frater J, Stöhr W, Hanke T, Fidler S, Dorrell L. Therapeutic vaccination following early antiretroviral therapy elicits highly functional T cell responses against conserved HIV-1 regions. Sci Rep 2023; 13:17155. [PMID: 37821472 PMCID: PMC10567821 DOI: 10.1038/s41598-023-42888-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
'Kick and kill' cure strategies aim to induce HIV protein expression in latently infected cells (kick), and thus trigger their elimination by cytolytic T cells (kill). In the Research in Viral Eradication of HIV Reservoirs trial (NCT02336074), people diagnosed with primary HIV infection received immediate antiretroviral therapy (ART) and were randomised 24 weeks later to either a latency-reversing agent, vorinostat, together with ChAdV63.HIVconsv and MVA.HIVconsv vaccines, or ART alone. This intervention conferred no reduction in HIV-1 reservoir size over ART alone, despite boosting virus-specific CD4+ and CD8+ T cells. The effects of the intervention were examined at the cellular level in the two trial arms using unbiased computational analysis of polyfunctional scores. This showed that the frequency and polyfunctionality of virus-specific CD4+ and CD8+ T cell populations were significantly increased over 12 weeks post-vaccination, compared to the ART-only arm. HIV-specific IL-2-secreting CD8+ T cells also expanded significantly in the intervention arm and were correlated with antiviral activity against heterologous HIV in vitro. Therapeutic vaccination during ART commenced in primary infection can induce functional T cell responses that are phenotypically similar to those of HIV controllers. Analytical therapy interruption may help determine their ability to control HIV in vivo.
Collapse
Affiliation(s)
- Jakub Kopycinski
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hongbing Yang
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gemma Hancock
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Matthew Pace
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ellen Kim
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Wolfgang Stöhr
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Tomás Hanke
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Joint Research Centre for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, and National Institute for Health Research Imperial Biomedical Research Centre, London, UK
| | - Lucy Dorrell
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Immunocore Ltd, 93 Park Drive, Milton Park, Abingdon, OX14 4RY, Oxon, UK.
| |
Collapse
|
42
|
Gunst JD, Højen JF, Pahus MH, Rosás-Umbert M, Stiksrud B, McMahon JH, Denton PW, Nielsen H, Johansen IS, Benfield T, Leth S, Gerstoft J, Østergaard L, Schleimann MH, Olesen R, Støvring H, Vibholm L, Weis N, Dyrhol-Riise AM, Pedersen KBH, Lau JSY, Copertino DC, Linden N, Huynh TT, Ramos V, Jones RB, Lewin SR, Tolstrup M, Rasmussen TA, Nussenzweig MC, Caskey M, Reikvam DH, Søgaard OS. Impact of a TLR9 agonist and broadly neutralizing antibodies on HIV-1 persistence: the randomized phase 2a TITAN trial. Nat Med 2023; 29:2547-2558. [PMID: 37696935 PMCID: PMC10579101 DOI: 10.1038/s41591-023-02547-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023]
Abstract
Inducing antiretroviral therapy (ART)-free virological control is a critical step toward a human immunodeficiency virus type 1 (HIV-1) cure. In this phase 2a, placebo-controlled, double-blinded trial, 43 people (85% males) with HIV-1 on ART were randomized to (1) placebo/placebo, (2) lefitolimod (TLR9 agonist)/placebo, (3) placebo/broadly neutralizing anti-HIV-1 antibodies (bNAbs) or (4) lefitolimod/bNAb. ART interruption (ATI) started at week 3. Lefitolimod was administered once weekly for the first 8 weeks, and bNAbs were administered twice, 1 d before and 3 weeks after ATI. The primary endpoint was time to loss of virologic control after ATI. The median delay in time to loss of virologic control compared to the placebo/placebo group was 0.5 weeks (P = 0.49), 12.5 weeks (P = 0.003) and 9.5 weeks (P = 0.004) in the lefitolimod/placebo, placebo/bNAb and lefitolimod/bNAb groups, respectively. Among secondary endpoints, viral doubling time was slower for bNAb groups compared to non-bNAb groups, and the interventions were overall safe. We observed no added benefit of lefitolimod. Despite subtherapeutic plasma bNAb levels, 36% (4/11) in the placebo/bNAb group compared to 0% (0/10) in the placebo/placebo group maintained virologic control after the 25-week ATI. Although immunotherapy with lefitolimod did not lead to ART-free HIV-1 control, bNAbs may be important components in future HIV-1 curative strategies. ClinicalTrials.gov identifier: NCT03837756 .
Collapse
Affiliation(s)
- Jesper D Gunst
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper F Højen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Marie H Pahus
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Miriam Rosás-Umbert
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Stiksrud
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - James H McMahon
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Paul W Denton
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Isik S Johansen
- Department of Infectious Diseases, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Leth
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Internal Medicine, Gødstrup Hospital, Gødstrup, Denmark
| | - Jan Gerstoft
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Lars Østergaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Mariane H Schleimann
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rikke Olesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik Støvring
- Department of Public Health, Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Line Vibholm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne M Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Karen B H Pedersen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jillian S Y Lau
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Dennis C Copertino
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Noemi Linden
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Tan T Huynh
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Victor Ramos
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - R Brad Jones
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sharon R Lewin
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas A Rasmussen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Dag Henrik Reikvam
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole S Søgaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
43
|
Harwood OE, Matschke LM, Moriarty RV, Balgeman AJ, Weaver AJ, Ellis-Connell AL, Weiler AM, Winchester LC, Fletcher CV, Friedrich TC, Keele BF, O’Connor DH, Lang JD, Reynolds MR, O’Connor SL. CD8+ cells and small viral reservoirs facilitate post-ART control of SIV replication in M3+ Mauritian cynomolgus macaques initiated on ART two weeks post-infection. PLoS Pathog 2023; 19:e1011676. [PMID: 37747933 PMCID: PMC10553806 DOI: 10.1371/journal.ppat.1011676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/05/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023] Open
Abstract
Sustainable HIV remission after antiretroviral therapy (ART) withdrawal, or post-treatment control (PTC), remains a top priority for HIV treatment. We observed surprising PTC in an MHC-haplomatched cohort of MHC-M3+ SIVmac239+ Mauritian cynomolgus macaques (MCMs) initiated on ART at two weeks post-infection (wpi). None of the MCMs possessed MHC haplotypes previously associated with SIV control. For six months after ART withdrawal, we observed undetectable or transient viremia in seven of the eight MCMs, despite detecting replication competent SIV using quantitative viral outgrowth assays. In vivo depletion of CD8α+ cells induced rebound in all animals, indicating the observed PTC was mediated, at least in part, by CD8α+ cells. With intact proviral DNA assays, we found that MCMs had significantly smaller viral reservoirs two wpi than a cohort of identically infected rhesus macaques, a population that rarely develops PTC. We found a similarly small viral reservoir among six additional SIV+ MCMs in which ART was initiated at eight wpi, some of whom exhibited viral rebound. These results suggest that an unusually small viral reservoir is a hallmark among SIV+ MCMs. By evaluating immunological differences between MCMs that did and did not rebound, we identified that PTC was associated with a reduced frequency of CD4+ and CD8+ lymphocyte subsets expressing exhaustion markers. Together, these results suggest a combination of small reservoirs and immune-mediated virus suppression contribute to PTC in MCMs. Further, defining the immunologic mechanisms that engender PTC in this model may identify therapeutic targets for inducing durable HIV remission in humans.
Collapse
Affiliation(s)
- Olivia E. Harwood
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Lea M. Matschke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ryan V. Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail J. Weaver
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Lee C. Winchester
- College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Courtney V. Fletcher
- College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Jessica D. Lang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Matthew R. Reynolds
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| |
Collapse
|
44
|
Bekker LG, Beyrer C, Mgodi N, Lewin SR, Delany-Moretlwe S, Taiwo B, Masters MC, Lazarus JV. HIV infection. Nat Rev Dis Primers 2023; 9:42. [PMID: 37591865 DOI: 10.1038/s41572-023-00452-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 08/19/2023]
Abstract
The AIDS epidemic has been a global public health issue for more than 40 years and has resulted in ~40 million deaths. AIDS is caused by the retrovirus, HIV-1, which is transmitted via body fluids and secretions. After infection, the virus invades host cells by attaching to CD4 receptors and thereafter one of two major chemokine coreceptors, CCR5 or CXCR4, destroying the host cell, most often a T lymphocyte, as it replicates. If unchecked this can lead to an immune-deficient state and demise over a period of ~2-10 years. The discovery and global roll-out of rapid diagnostics and effective antiretroviral therapy led to a large reduction in mortality and morbidity and to an expanding group of individuals requiring lifelong viral suppressive therapy. Viral suppression eliminates sexual transmission of the virus and greatly improves health outcomes. HIV infection, although still stigmatized, is now a chronic and manageable condition. Ultimate epidemic control will require prevention and treatment to be made available, affordable and accessible for all. Furthermore, the focus should be heavily oriented towards long-term well-being, care for multimorbidity and good quality of life. Intense research efforts continue for therapeutic and/or preventive vaccines, novel immunotherapies and a cure.
Collapse
Affiliation(s)
- Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, RSA, Cape Town, South Africa.
| | - Chris Beyrer
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Nyaradzo Mgodi
- University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | | | - Babafemi Taiwo
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Mary Clare Masters
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Jeffrey V Lazarus
- CUNY Graduate School of Public Health and Health Policy, New York, NY, USA
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
45
|
Arenas VR, Rugeles MT, Perdomo-Celis F, Taborda N. Recent advances in CD8 + T cell-based immune therapies for HIV cure. Heliyon 2023; 9:e17481. [PMID: 37441388 PMCID: PMC10333625 DOI: 10.1016/j.heliyon.2023.e17481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Achieving a cure for HIV infection is a global priority. There is substantial evidence supporting a central role for CD8+ T cells in the natural control of HIV, suggesting the rationale that these cells may be exploited to achieve remission or cure of this infection. In this work, we review the major challenges for achieving an HIV cure, the models of HIV remission, and the mechanisms of HIV control mediated by CD8+ T cells. In addition, we discuss strategies based on this cell population that could be used in the search for an HIV cure. Finally, we analyze the current challenges and perspectives to translate this basic knowledge toward scalable HIV cure strategies.
Collapse
Affiliation(s)
| | - María T. Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | | | - Natalia Taborda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellin, Colombia
| |
Collapse
|
46
|
Vlaming KE, van Wijnbergen K, Kaptein TM, Nijhuis M, Kootstra NJ, de Bree GJ, Geijtenbeek TB. Crosstalk between TLR8 and RIG-I-like receptors enhances antiviral immune responses. Front Med (Lausanne) 2023; 10:1146457. [PMID: 37261119 PMCID: PMC10227620 DOI: 10.3389/fmed.2023.1146457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
Background Toll-like receptor (TLR) agonists have been investigated due to their potential dual effects as latency reverting agents and immune modulatory compounds in people living with HIV (PLWH). Here, we investigated whether co-stimulation of TLR7/8 agonists with RIG-I-like receptor (RLR) agonists enhances antiviral immunity. Methods Peripheral blood mononuclear cells (PBMCs) and monocyte-derived dendritic cells (DCs) were incubated with TLR and RLR-agonists for 24 h and innate and adaptive immune responses were determined (maturation markers, cytokines in supernatant, ISG expression). Results Both TLR7 and TLR8 agonists induced pro-inflammatory cytokines in DCs as well as PBMCs. TLR8 agonists were more potent in inducing cytokine responses and had a stronger effect on DC-induced immunity. Notably, while all compounds induced IL-12p70, co-stimulation with TLR8 agonists and RLR agonist polyI: C induced significantly higher levels of IL-12p70 in PBMCs. Moreover, crosstalk between TLR8 and RLR agonists induced a strong type I Interferon (IFN) response as different antiviral IFN-stimulated genes were upregulated by the combination compared to the agonists alone. Conclusion Our data strongly suggest that TLR crosstalk with RLRs leads to strong antiviral immunity as shown by induction of IL-12 and type I IFN responses in contrast to TLRs alone. Thus, co-stimulation of TLRs and RLRs might be a powerful strategy to induce reactivation of latent reservoir as well as antiviral immunity that eliminates the reactivated cells.
Collapse
Affiliation(s)
- Killian E. Vlaming
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Kelly van Wijnbergen
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Tanja M. Kaptein
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Monique Nijhuis
- Translational Virology, Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Neeltje J. Kootstra
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Godelieve J. de Bree
- Department of Internal Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Teunis B. Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| |
Collapse
|
47
|
Esmaeilzadeh E, Etemad B, Lavine CL, Garneau L, Li Y, Regan J, Wong C, Sharaf R, Connick E, Volberding P, Sagar M, Seaman MS, Li JZ. Autologous neutralizing antibodies increase with early antiretroviral therapy and shape HIV rebound after treatment interruption. Sci Transl Med 2023; 15:eabq4490. [PMID: 37163616 PMCID: PMC10576978 DOI: 10.1126/scitranslmed.abq4490] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/10/2023] [Indexed: 05/12/2023]
Abstract
Early initiation of antiretroviral therapy (ART) alters viral rebound kinetics after analytic treatment interruption (ATI) and may play a role in promoting HIV-1 remission. Autologous neutralizing antibodies (aNAbs) represent a key adaptive immune response in people living with HIV-1. We aimed to investigate the role of aNAbs in shaping post-ATI HIV-1 rebound variants. We performed single-genome amplification of HIV-1 env from pre-ART and post-ATI plasma samples of 12 individuals who initiated ART early after infection. aNAb activity was quantified using pseudoviruses derived from the most common plasma variant, and the serum dilution that inhibited 50% of viral infections was determined. aNAb responses matured while participants were on suppressive ART, because on-ART plasma and purified immunoglobulin G (IgG) demonstrated improved neutralizing activity against pre-ART HIV-1 strains when compared with pre-ART plasma or purified IgG. Post-ATI aNAb responses exerted selective pressure on the rebounding viruses, because the post-ATI HIV-1 strains were more resistant to post-ATI plasma neutralization compared with the pre-ART virus. Several pre-ATI features distinguished post-treatment controllers from noncontrollers, including an infecting HIV-1 sequence that was more similar to consensus HIV-1 subtype B, more restricted proviral diversity, and a stronger aNAb response. Post-treatment control was also associated with the evolution of distinct N-glycosylation profiles in the HIV-1 envelope. In summary, aNAb responses appeared to mature after early initiation of ART and applied selective pressure on rebounding viruses. The combination of aNAb activity with select HIV-1 sequence and reservoir features identified individuals with a greater chance of post-treatment control.
Collapse
Affiliation(s)
| | - Behzad Etemad
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Lauren Garneau
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Yijia Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James Regan
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Colline Wong
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Radwa Sharaf
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Paul Volberding
- University of California, San Francisco, San Francisco, CA 94158, USA
| | - Manish Sagar
- Boston University Chobanian and Avedesian School of Medicine, Boston, MA 02118, USA
| | | | - Jonathan Z Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Zhou C, Wu Y, Zhang Y, Wang Y, Wu H, Zhang T, Chen G, Huang X. Factors associated with post-treatment control of viral load in HIV-infected patients: a systematic review and meta-analysis. Int J Infect Dis 2023; 129:216-227. [PMID: 36707043 DOI: 10.1016/j.ijid.2023.01.025] [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: 08/16/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVES This study aimed to investigate the factors associated with maintenance of viral suppression after antiretroviral therapy (ART) discontinuation. METHODS Databases were searched for studies published between January 01, 2011, and July 01, 2022, that correlated the time of virus rebound with treatment interruption (TI). The corresponding data were extracted from these studies. A fixed-effects model was used to calculate pooled estimates. RESULTS Thirty-one studies were included in this analysis. Results showed that patients who started ART during acute or early infection had longer viral control than those who started ART during chronic infection. It has been reported that some broadly neutralizing HIV-1-specific antibodies can significantly prolong viral inhibition. The study also found that approximately 7.2% of patients achieved post-treatment control (PTC) approximately a year after TI. CONCLUSION ART initiation in the acute or early phases can delay viral rebound after TI. Cell-associated HIV RNA and HIV DNA have been difficult to prove as able to predict viral rebound time. Many vaccines and antibodies have also been shown to be effective in prolonging viral control in people without PTC, and more research is needed to develop alternative ART therapies that can effectively inhibit or even eliminate HIV.
Collapse
Affiliation(s)
- Chi Zhou
- Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao, China; Clinical and Research Center for Infectious Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yaxin Wu
- Clinical and Research Center for Infectious Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Clinical and Research Center for Infectious Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yingying Wang
- Department of Internal Medicine, Shenzhen Hospital of the University of Hong Kong, Shenzhen, China
| | - Hao Wu
- Clinical and Research Center for Infectious Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Clinical and Research Center for Infectious Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| | - Guanzhi Chen
- Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xiaojie Huang
- Clinical and Research Center for Infectious Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
49
|
Etemad B, Sun X, Li Y, Melberg M, Moisi D, Gottlieb R, Ahmed H, Aga E, Bosch RJ, Acosta EP, Yuki Y, Martin MP, Carrington M, Gandhi RT, Jacobson JM, Volberding P, Connick E, Mitsuyasu R, Frank I, Saag M, Eron JJ, Skiest D, Margolis DM, Havlir D, Schooley RT, Lederman MM, Yu XG, Li JZ. HIV post-treatment controllers have distinct immunological and virological features. Proc Natl Acad Sci U S A 2023; 120:e2218960120. [PMID: 36877848 PMCID: PMC10089217 DOI: 10.1073/pnas.2218960120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023] Open
Abstract
HIV post-treatment controllers (PTCs) are rare individuals who maintain low levels of viremia after stopping antiretroviral therapy (ART). Understanding the mechanisms of HIV post-treatment control will inform development of strategies aiming at achieving HIV functional cure. In this study, we evaluated 22 PTCs from 8 AIDS Clinical Trials Group (ACTG) analytical treatment interruption (ATI) studies who maintained viral loads ≤400 copies/mL for ≥24 wk. There were no significant differences in demographics or frequency of protective and susceptible human leukocyte antigen (HLA) alleles between PTCs and post-treatment noncontrollers (NCs, n = 37). Unlike NCs, PTCs demonstrated a stable HIV reservoir measured by cell-associated RNA (CA-RNA) and intact proviral DNA assay (IPDA) during analytical treatment interruption (ATI). Immunologically, PTCs demonstrated significantly lower CD4+ and CD8+ T cell activation, lower CD4+ T cell exhaustion, and more robust Gag-specific CD4+ T cell responses and natural killer (NK) cell responses. Sparse partial least squares discriminant analysis (sPLS-DA) identified a set of features enriched in PTCs, including a higher CD4+ T cell% and CD4+/CD8+ ratio, more functional NK cells, and a lower CD4+ T cell exhaustion level. These results provide insights into the key viral reservoir features and immunological profiles for HIV PTCs and have implications for future studies evaluating interventions to achieve an HIV functional cure.
Collapse
Affiliation(s)
- Behzad Etemad
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Xiaoming Sun
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
| | - Yijia Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Meghan Melberg
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Daniela Moisi
- School of Medicine, Case Western Reserve University, Cleveland, OH44106
| | - Rachel Gottlieb
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Hayat Ahmed
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Evgenia Aga
- Harvard T. H. Chan School of Public Health, Boston, MA02115
| | | | - Edward P. Acosta
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL35233
| | - Yuko Yuki
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD21702
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD20814
| | - Maureen P. Martin
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD21702
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD20814
| | - Mary Carrington
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD21702
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD20814
| | - Rajesh T. Gandhi
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | | | - Paul Volberding
- School of Medicine, University of California San Francisco, San Francisco, CA94143
| | | | - Ronald Mitsuyasu
- School of Medicine, University of California Los Angeles, Los Angeles, CA90095
| | - Ian Frank
- School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michael Saag
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL35233
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Daniel Skiest
- Department of Medicine, University of Massachusetts Chan Medical School - Baystate, Springfield, MA01199
| | - David M. Margolis
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Diane Havlir
- School of Medicine, University of California San Francisco, San Francisco, CA94143
| | - Robert T. Schooley
- Department of Medicine, University of California San Diego, San Diego, CA92103
| | | | - Xu G. Yu
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
| | - Jonathan Z. Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| |
Collapse
|
50
|
Rodríguez-Agustín A, Casanova V, Grau-Expósito J, Sánchez-Palomino S, Alcamí J, Climent N. Immunomodulatory Activity of the Tyrosine Kinase Inhibitor Dasatinib to Elicit NK Cytotoxicity against Cancer, HIV Infection and Aging. Pharmaceutics 2023; 15:pharmaceutics15030917. [PMID: 36986778 PMCID: PMC10055786 DOI: 10.3390/pharmaceutics15030917] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have been extensively used as a treatment for chronic myeloid leukemia (CML). Dasatinib is a broad-spectrum TKI with off-target effects that give it an immunomodulatory capacity resulting in increased innate immune responses against cancerous cells and viral infected cells. Several studies reported that dasatinib expanded memory-like natural killer (NK) cells and γδ T cells that have been related with increased control of CML after treatment withdrawal. In the HIV infection setting, these innate cells are associated with virus control and protection, suggesting that dasatinib could have a potential role in improving both the CML and HIV outcomes. Moreover, dasatinib could also directly induce apoptosis of senescence cells, being a new potential senolytic drug. Here, we review in depth the current knowledge of virological and immunogenetic factors associated with the development of powerful cytotoxic responses associated with this drug. Besides, we will discuss the potential therapeutic role against CML, HIV infection and aging.
Collapse
Affiliation(s)
| | - Víctor Casanova
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Judith Grau-Expósito
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Sonsoles Sánchez-Palomino
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC), 28029 Madrid, Spain
| | - José Alcamí
- CIBER of Infectious Diseases (CIBERINFEC), 28029 Madrid, Spain
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Núria Climent
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-93-2275400 (ext. 3144); Fax: +34-93-2271775
| |
Collapse
|