1
|
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
|
2
|
Nel C, Frater J. Enhancing broadly neutralising antibody suppression of HIV by immune modulation and vaccination. Front Immunol 2024; 15:1478703. [PMID: 39575236 PMCID: PMC11578998 DOI: 10.3389/fimmu.2024.1478703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Although HIV infection can be managed with antiretroviral drugs, there is no cure and therapy has to be taken for life. Recent successes in animal models with HIV-specific broadly neutralising antibodies (bNAbs) have led to long-term virological remission and even possible cures in some cases. This has resulted in substantial investment in human studies to explore bNAbs as a curative intervention for HIV infection. Emerging data are encouraging, but suggest that combinations of bNAbs with other immunomodulatory agents may be needed to induce and sustain long-term viral control. As a result, a number of clinical trials are currently underway exploring these combinations. If successful, the impact for the millions of people living with HIV could be substantial. Here, we review the background to the use of bNAbs in the search for an HIV cure and how different adjunctive agents might be used together to enhance their efficacy.
Collapse
Affiliation(s)
- Carla Nel
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- The National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
3
|
Manning MR, Blazkova J, Justement JS, Shi V, Kennedy BD, Rai MA, Seamon CA, Gittens K, Sneller MC, Moir S, Chun TW. Timing of antiretroviral therapy initiation affects intact HIV reservoirs following analytical treatment interruption. J Clin Invest 2024; 134:e181632. [PMID: 39403920 PMCID: PMC11473155 DOI: 10.1172/jci181632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Affiliation(s)
- Maegan R. Manning
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Jana Blazkova
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Jesse S. Justement
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Victoria Shi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Brooke D. Kennedy
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - M. Ali Rai
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Catherine A. Seamon
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA
| | - Kathleen Gittens
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA
| | - Michael C. Sneller
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases and
| |
Collapse
|
4
|
Alagaratnam J, Stöhr W, Hamlyn E, Porter K, Toombs J, Heslegrave A, Zetterberg H, Gisslén M, Underwood J, Schechter M, Kaleebu P, Tambussi G, Kinloch S, Miro JM, Kelleher AD, Babiker A, Frater J, Winston A, Fidler S. Impact of interrupting antiretroviral therapy started during primary HIV-1 infection on plasma neurofilament light chain protein, a marker of neuronal injury: The SPARTAC trial. J Virus Erad 2024; 10:100381. [PMID: 38988673 PMCID: PMC11234014 DOI: 10.1016/j.jve.2024.100381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Objective Antiretroviral therapy (ART)-conferred suppression of HIV replication limits neuronal injury and inflammation. ART interruption tests efficacy in HIV cure trials and viral rebound after ART interruption may induce neuronal injury. We investigated the impact of protocol-defined ART interruption, commenced during primary HIV-1 infection (PHI) on a biomarker of neuro-axonal injury (neurofilament light protein (NfL)), and its associations with inflammation (D-dimer and interleukin-6 (IL-6)) and HIV-1 reservoir size (total HIV-1 DNA). Design Retrospective study measuring plasma NfL in 83 participants enrolled in SPARTAC randomised to receive 48-weeks ART initiated during PHI, followed by ART interruption. Methods NfL (Simoa immunoassay, Quanterix™) was measured before ART, after 48 weeks on ART, and 12 weeks after stopping ART. Plasma D-dimer and IL-6, and total HIV-1 DNA in peripheral CD4+ T-cells results were available in a subset of participants. Longitudinal NfL changes were assessed using mixed models, and associations with clinical and laboratory parameters using linear regression. Results NfL decreased following 48-weeks ART (geometric mean 6.9 to 5.8 pg/mL, p = 0.006) with no further significant change up to 12-weeks post-stopping ART despite viral rebound in the majority of participants (median 1.7 to 3.9 plasma HIV-1 RNA log10 copies/mL). Higher baseline NfL was independently associated with higher plasma HIV-1 RNA (p = 0.020) and older age (p = 0.002). While NfL was positively associated with D-dimer (n = 48; p = 0.002), there was no significant association with IL-6 (n = 48) or total HIV-1 DNA (n = 51). Conclusions Using plasma NfL as a surrogate marker, a decrease in neuro-axonal injury was observed in a cohort of participants following ART initiation during PHI, with no evidence of neuro-axonal injury rebound following ART interruption for up to 12 weeks, despite viral rebound in the majority of participants.
Collapse
Affiliation(s)
- Jasmini Alagaratnam
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Genitourinary Medicine/ HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Wolfgang Stöhr
- Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
| | - Elizabeth Hamlyn
- Caldecot Centre, Kings College Hospital NHS Foundation Trust, London, United Kingdom
| | - Kholoud Porter
- Institute for Global Health, University College London, London, United Kingdom
| | - Jamie Toombs
- UK Dementia Research Institute at University College London, London, United Kingdom
| | - Amanda Heslegrave
- UK Dementia Research Institute at University College London, London, United Kingdom
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Diseases, Gothenburg, Sweden
| | - Jonathan Underwood
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mauro Schechter
- Projeto Praça Onze, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pontiano Kaleebu
- Medical Research Council/Uganda Virus Research Institute, Entebbe, Uganda
| | | | - Sabine Kinloch
- Department of Infection and Immunity, Royal Free Hospital, Pond Street, London, United Kingdom
| | - Jose M Miro
- Infectious Diseases Service, Hospital Clinic - IDIBAPS. University of Barcelona, Barcelona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Abdel Babiker
- Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute of Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Alan Winston
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Genitourinary Medicine/ HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Sarah Fidler
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- Genitourinary Medicine/ HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
5
|
Kumar MR, Fray EJ, Bender AM, Zitzmann C, Ribeiro RM, Perelson AS, Barouch DH, Siliciano JD, Siliciano RF. Biphasic decay of intact SHIV genomes following initiation of antiretroviral therapy complicates analysis of interventions targeting the reservoir. Proc Natl Acad Sci U S A 2023; 120:e2313209120. [PMID: 37844236 PMCID: PMC10614214 DOI: 10.1073/pnas.2313209120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/05/2023] [Indexed: 10/18/2023] Open
Abstract
The latent reservoir for HIV-1 in resting CD4+ T cells persists despite antiretroviral therapy (ART) and precludes cure. Reservoir-targeting interventions are evaluated in ART-treated macaques infected with simian immunodeficiency virus (SIV) or simian-human immunodeficiency virus (SHIV). Efficacy is determined by reservoir measurements before and after the intervention. However, most proviruses persisting in the setting of ART are defective. In addition, intact HIV-1 and SIV genomes undergo complex, multiphasic decay observable when new infection events are blocked by ART. Intervention-induced elimination of latently infected cells must be distinguished from natural decay. Here, we address these issues for SHIV. We describe an intact proviral DNA assay that allows digital counting of SHIV genomes lacking common fatal defects. We show that intact SHIV genomes in circulating CD4+ T cells undergo biphasic decay during the first year of ART, with a rapid first phase (t1/2 = 30.1 d) and a slower second phase (t1/2 = 8.1 mo) that is still more rapid that the slow decay observed in people with HIV-1 on long-term ART (t1/2 = 3.7 y). In SHIV models, most interventions are tested during 2nd phase decay. Natural 2nd phase decay must be considered in evaluating interventions as most infected cells present at this time do not become part of the stable reservoir. In addition, for interventions tested during 2nd phase decay, a caveat is that the intervention may not be equally effective in people with HIV on long-term ART whose reservoirs are dominated by latently infected cells with a slower decay rate.
Collapse
Affiliation(s)
- Mithra R. Kumar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Emily J. Fray
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Alexandra M. Bender
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | | | | | | | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA02215
| | - Janet D. Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Robert F. Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
- HHMI, Baltimore, MD21205
| |
Collapse
|
6
|
Dubé K, Campbell CK, Eskaf S, Sauceda JA, Ndukwe S, Henley L, Persaud D, Deeks SG, Auerbach JD, Saberi P. Willingness of Racially Diverse Young Adults Living with HIV to Participate in HIV Cure Research: A Cross-Sectional Survey in the United States. AIDS Res Hum Retroviruses 2023; 39:381-399. [PMID: 36226414 PMCID: PMC10387162 DOI: 10.1089/aid.2022.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nearly half of new HIV cases in the United States are among youth. Little is known about the willingness of young adults living with HIV (YLWH) to participate in HIV cure-related research. In 2021, we recruited 271 YLWH aged 18-29 for an online survey. We asked questions about willingness to participate in HIV cure research, perceived risks and benefits, acceptable trade-offs, and perceptions on analytical treatment interruptions. We conducted descriptive analyses to summarize data and bivariate analyses to explore correlations by demographics. Most respondents (mean age = 26) identified as men (86%) and Black Americans (69%). YLWH expressed high willingness to consider participating in cell- and gene-based approaches (75%) and immune-based approaches (71%). Approximately 45% would be willing to let their viral load become detectable for a period of time during an HIV cure study, 27% would not be willing, and 28% did not know. The social risk most likely to deter participation was the possibility of transmitting HIV to sex partners while off HIV medications (65% of respondents would be deterred a great deal or a lot). Compared to the 25-29 age group (n = 192), the 18-24 age group (n = 79) was more likely to indicate that having to disclose HIV status would matter a great deal in considering participation in HIV cure research (38% vs. 21%, p = .003). Inclusion and engagement of YLWH are critical for advancing novel HIV curative agents. Our article concludes with possible considerations for engaging YLWH in HIV cure research. Physical, clinical, and social risks will need to be kept to a minimum, and research teams will need to proactively mitigate the possibility of transmitting HIV to sex partners while off HIV medications.
Collapse
Affiliation(s)
- Karine Dubé
- Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Chadwick K. Campbell
- Department of Medicine, Center for AIDS Prevention Studies (CAPS), University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Shadi Eskaf
- Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - John A. Sauceda
- Department of Medicine, Center for AIDS Prevention Studies (CAPS), University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Samuel Ndukwe
- Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Laney Henley
- Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Deborah Persaud
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven G. Deeks
- Department of Medicine, Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco (UCSF). San Francisco, California, USA
| | - Judith D. Auerbach
- Department of Medicine, Center for AIDS Prevention Studies (CAPS), University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Parya Saberi
- Department of Medicine, Center for AIDS Prevention Studies (CAPS), University of California, San Francisco (UCSF), San Francisco, California, USA
| |
Collapse
|
7
|
Scully EP, Aga E, Tsibris A, Archin N, Starr K, Ma Q, Morse GD, Squires KE, Howell BJ, Wu G, Hosey L, Sieg SF, Ehui L, Giguel F, Coxen K, Dobrowolski C, Gandhi M, Deeks S, Chomont N, Connick E, Godfrey C, Karn J, Kuritzkes DR, Bosch RJ, Gandhi RT. Impact of Tamoxifen on Vorinostat-Induced Human Immunodeficiency Virus Expression in Women on Antiretroviral Therapy: AIDS Clinical Trials Group A5366, The MOXIE Trial. Clin Infect Dis 2022; 75:1389-1396. [PMID: 35176755 PMCID: PMC9555843 DOI: 10.1093/cid/ciac136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Biological sex and the estrogen receptor alpha (ESR1) modulate human immunodeficiency virus (HIV) activity. Few women have enrolled in clinical trials of latency reversal agents (LRAs); their effectiveness in women is unknown. We hypothesized that ESR1 antagonism would augment induction of HIV expression by the LRA vorinostat. METHODS AIDS Clinical Trials Group A5366 enrolled 31 virologically suppressed, postmenopausal women on antiretroviral therapy. Participants were randomized 2:1 to receive tamoxifen (arm A, TAMOX/VOR) or observation (arm B, VOR) for 5 weeks followed by 2 doses of vorinostat. Primary end points were safety and the difference between arms in HIV RNA induction after vorinostat. Secondary analyses included histone 4 acetylation, HIV DNA, and plasma viremia by single copy assay (SCA). RESULTS No significant adverse events were attributed to study treatments. Tamoxifen did not enhance vorinostat-induced HIV transcription (between-arm ratio, 0.8; 95% confidence interval [CI], .2-2.4). Vorinostat-induced HIV transcription was higher in participants with increases in H4Ac (fold increase, 2.78; 95% CI, 1.34-5.79) vs those 9 who did not (fold increase, 1.04; 95% CI, .25-4.29). HIV DNA and SCA plasma viremia did not substantially change. CONCLUSIONS Tamoxifen did not augment vorinostat-induced HIV RNA expression in postmenopausal women. The modest latency reversal activity of vorinostat, postmenopausal status, and low level of HIV RNA expression near the limits of quantification limited assessment of the impact of tamoxifen. This study is the first HIV cure trial done exclusively in women and establishes both the feasibility and necessity of investigating novel HIV cure strategies in women living with HIV. CLINICAL TRIALS REGISTRATION NCT03382834.
Collapse
Affiliation(s)
- Eileen P Scully
- Departement of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Evgenia Aga
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Athe Tsibris
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nancie Archin
- University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kate Starr
- ACTG Clinical Research Site, Ohio State University, Hilliard, Ohio, USA
| | - Qing Ma
- Translational Pharmacology Research Core, University at Buffalo, Buffalo, New York, USA
| | - Gene D Morse
- Translational Pharmacology Research Core, University at Buffalo, Buffalo, New York, USA
| | | | - Bonnie J Howell
- Department of Infectious Disease and Vaccines, Merck and Co, West Point, Pennsylvania, USA
| | - Guoxin Wu
- Department of Infectious Disease and Vaccines, Merck and Co, West Point, Pennsylvania, USA
| | - Lara Hosey
- ACTG Network Coordinating Center, Silver Spring, Maryland, USA
| | - Scott F Sieg
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Lynsay Ehui
- Whitman-Walker Health, Washington, D.C., USA
| | - Francoise Giguel
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kendyll Coxen
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Curtis Dobrowolski
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Monica Gandhi
- Department of Medicine, University of California, San Francisco, California, USA
| | - Steve Deeks
- Department of Medicine, University of California, San Francisco, California, USA
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Canada
| | | | - Catherine Godfrey
- Office of the Global AIDS Coordinator, Department of State, Washington D.C., USA
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Daniel R Kuritzkes
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronald J Bosch
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Rajesh T Gandhi
- Department of Medicine, Massachusetts General Hospital, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Adams P, Berkhout B, Pasternak AO. Towards a molecular profile of antiretroviral therapy-free HIV remission. Curr Opin HIV AIDS 2022; 17:301-307. [PMID: 35938464 DOI: 10.1097/coh.0000000000000749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To summarize the current status and highlight recent findings on predictive biomarkers for posttreatment HIV control (PTC) and virological remission. While historically, many studies focused on virological markers, there is an increasing tendency to enter immune and metabolic factors into the equation. RECENT FINDINGS On the virological side, several groups reported that cell-associated HIV RNA could predict time to viral rebound. Recent data hints at the possible importance of the genic location and chromatin context of the integrated provirus, although these factors still need to be assessed in relation to PTC and virological remission. Evidence from immunological studies highlighted innate and humoral immunity as important factors for prolonged HIV remission. Interestingly, novel metabolic markers have emerged, which offer additional angles to our understanding of latency and viral rebound. SUMMARY Facilitating PTC and virological remission remain top priorities for the HIV cure research. We advocate for clear and precise definitions for both phenomena in order to avoid misconceptions and to strengthen the conclusions that can be drawn. As no one-size-fits-all marker has emerged yet, more biomarkers are on the horizon, and viral rebound is a complex and heterogeneous process, it is likely that a combination of various biomarkers in cohesion will be necessary for a more accurate prediction of antiretroviral therapy-free HIV remission.
Collapse
Affiliation(s)
- Philipp Adams
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
9
|
Bocharov G, Grebennikov D, Cebollada Rica P, Domenjo-Vila E, Casella V, Meyerhans A. Functional cure of a chronic virus infection by shifting the virus - host equilibrium state. Front Immunol 2022; 13:904342. [PMID: 36110838 PMCID: PMC9468810 DOI: 10.3389/fimmu.2022.904342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical handling of chronic virus infections remains a challenge. Here we describe recent progress in the understanding of virus - host interaction dynamics. Based on the systems biology concept of multi-stability and the prediction of multiplicative cooperativity between virus-specific cytotoxic T cells and neutralising antibodies, we argue for the requirements to engage multiple immune system components for functional cure strategies. Our arguments are derived from LCMV model system studies and are translated to HIV-1 infection.
Collapse
Affiliation(s)
- Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow, Russia
- Moscow Center for Fundamental and Applied Mathematics at INM RAS, Moscow, Russia
- Institute for Computer Science and Mathematical Modelling, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dmitry Grebennikov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow, Russia
- Moscow Center for Fundamental and Applied Mathematics at INM RAS, Moscow, Russia
- Institute for Computer Science and Mathematical Modelling, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Paula Cebollada Rica
- Infection Biology Laboratory, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eva Domenjo-Vila
- Infection Biology Laboratory, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Valentina Casella
- Infection Biology Laboratory, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
10
|
Genotypic and Phenotypic Diversity of the Replication-Competent HIV Reservoir in Treated Patients. Microbiol Spectr 2022; 10:e0078422. [PMID: 35770985 PMCID: PMC9431663 DOI: 10.1128/spectrum.00784-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In HIV infection, viral rebound after treatment discontinuation is considered to originate predominantly from viral genomes integrated in resting CD4+ T lymphocytes. Replication-competent proviral genomes represent a minority of the total HIV DNA. While the quantification of the HIV reservoir has been extensively studied, the diversity of genomes that compose the reservoir was less explored. Here, we measured the genotypic and phenotypic diversity in eight patients with different treatment histories. Between 4 and 14 (mean, 8) individual viral isolates per patient were obtained using a virus outgrowth assay, and their near-full-length genomes were sequenced. The mean pairwise distance (MPD) observed in different patients correlated with the time before undetectable viremia was achieved (r = 0.864, P = 0.0194), suggesting that the complexity of the replication-competent reservoir mirrors that present at treatment initiation. No correlation was instead observed between MPD and the duration of successful treatment (mean, 8 years; range, 2 to 21 years). For 5 of the 8 patients, genotypically identical viral isolates were observed in independent wells, suggesting clonal expansion of infected cells. Identical viruses represented between 25 and 60% of the isolates (mean, 48%). The proportion of identical viral isolates correlated with the duration of treatment (r = 0.822, P = 0.0190), suggesting progressive clonal expansion of infected cells during ART. A broader range of infectivity was also observed among isolates from patients with delayed viremia control (r = 0.79, P = 0.025). This work unveiled differences in the genotypic and phenotypic features of the replication-competent reservoir from treated patients and suggests that delaying treatment results in increased diversity of the reservoir. IMPORTANCE In HIV-infected and effectively treated individuals, integrated proviral genomes may persist for decades. The vast majority of the genomes, however, are defective, and only the replication-competent fraction represents a threat of viral reemergence. The quantification of the reservoir has been thoroughly explored, while the diversity of the genomes has been insufficiently studied. Its characterization, however, is relevant for the design of strategies aiming the reduction of the reservoir. Here, we explored the replication-competent near-full-length HIV genomes of eight patients who experienced differences in the delay before viremia control and in treatment duration. We found that delayed effective treatment was associated with increased genetic diversity of the reservoir. The duration of treatment did not impact the diversity but was associated with higher frequency of clonally expanded sequences. Thus, early treatment initiation has the double advantage of reducing both the size and the diversity of the reservoir.
Collapse
|
11
|
Martínez-Román P, Crespo-Bermejo C, Valle-Millares D, Lara-Aguilar V, Arca-Lafuente S, Martín-Carbonero L, Ryan P, de los Santos I, López-Huertas MR, Palladino C, Muñoz-Muñoz M, Fernández-Rodríguez A, Coiras M, Briz V. Dynamics of HIV Reservoir and HIV-1 Viral Splicing in HCV-Exposed Individuals after Elimination with DAAs or Spontaneous Clearance. J Clin Med 2022; 11:3579. [PMID: 35806864 PMCID: PMC9267476 DOI: 10.3390/jcm11133579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Background: Although human immunodeficiency virus type 1 (HIV-1) reservoir size is very stable under antiretroviral therapy (ART), individuals exposed to the Hepatitis C virus (HCV) (chronically coinfected and spontaneous clarifiers) show an increase in HIV reservoir size and in spliced viral RNA, which could indicate that the viral protein regulator Tat is being more actively synthesized and, thus, could lead to a higher yield of new HIV. However, it is still unknown whether the effect of HCV elimination with direct-acting antivirals (DAAs) could modify the HIV reservoir and splicing. Methods: This longitudinal study (48 weeks’ follow-up after sustained virological response) involves 22 HIV+-monoinfected individuals, 17 HIV+/HCV- spontaneous clarifiers, and 24 HIV+/HCV+ chronically infected subjects who eliminated HCV with DAAs (all of them aviremic, viral load < 50). Viral-spliced RNA transcripts and proviral DNA copies were quantified by qPCR. Paired samples were analyzed using a mixed generalized linear model. Results: A decrease in HIV proviral DNA was observed in HIV+/HCV- subjects, but no significant differences were found for the other study groups. An increased production of multiple spliced transcripts was found in HIV+ and HIV+/HCV+ individuals. Conclusions: We conclude that elimination of HCV by DAAs was unable to revert the consequences derived from chronic HCV infection for the reservoir size and viral splicing, which could indicate an increased risk of rapid HIV-reservoir reactivation. Moreover, spontaneous clarifiers showed a significant decrease in the HIV reservoir, likely due to an enhanced immune response in these individuals.
Collapse
Affiliation(s)
- Paula Martínez-Román
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | - Celia Crespo-Bermejo
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | - Daniel Valle-Millares
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | - Violeta Lara-Aguilar
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | - Sonia Arca-Lafuente
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | - Luz Martín-Carbonero
- Instituto de Investigación Sanitaria Hospital de la Paz (IdiPAZ), 28046 Madrid, Spain;
| | - Pablo Ryan
- Department of Infectious Diseases, Infanta Leonor Hospital, 28031 Madrid, Spain;
| | - Ignacio de los Santos
- Servicio de Medicina Interna-Infecciosas, Hospital Universitario de La Princesa, 28006 Madrid, Spain;
| | - María Rosa López-Huertas
- Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (M.R.L.-H.); (M.C.)
| | - Claudia Palladino
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisbon, Portugal;
| | - María Muñoz-Muñoz
- Department of Animal Genetics, Instituto Nacional de Investigación y Tecnnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain;
| | - Amanda Fernández-Rodríguez
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | - Mayte Coiras
- Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (M.R.L.-H.); (M.C.)
| | - Verónica Briz
- Laboratory of Reference and Research on Viral Hepatitis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Madrid, Spain; (P.M.-R.); (C.C.-B.); (D.V.-M.); (V.L.-A.); (S.A.-L.)
| | | |
Collapse
|
12
|
Peay HL, Rennie S, Cadigan RJ, Gwaltney A, Jupimai T, Phanuphak N, Kroon E, Colby DJ, Ormsby N, Isaacson SC, Vasan S, Sacdalan C, Prueksakaew P, Benjapornpong K, Ananworanich J, Henderson GE. Attitudes About Analytic Treatment Interruption (ATI) in HIV Remission Trials with Different Antiretroviral Therapy (ART) Resumption Criteria. AIDS Behav 2022; 26:1504-1516. [PMID: 34997386 PMCID: PMC9007833 DOI: 10.1007/s10461-021-03504-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/01/2022]
Abstract
HIV remission trials often require temporary stopping of antiretroviral therapy (ART)-an approach called analytic treatment interruption (ATI). Trial designs resulting in viremia raise risks for participants and sexual partners. We conducted a survey on attitudes about remission trials, comparing ART resumption criteria (lower-risk "time to rebound" and higher-risk "sustained viremia") among participants from an acute HIV cohort in Thailand. Analyses included Wilcoxon-Ranks and multivariate logistic analysis. Most of 408 respondents supported ATI trials, with slightly higher approval of, and willingness to participate in, trials using time to rebound versus sustained viremia criteria. Less than half of respondents anticipated disclosing trial participation to partners and over half indicated uncertainty or unwillingness about whether partners would be willing to use PrEP. Willingness to participate was higher among those who rated higher trial approval, lower anticipated burden, and those expecting to make the decision independently. Our findings support acceptability of ATI trials among most respondents. Participant attitudes and anticipated behaviors, especially related to transmission risk, have implications for future trial design and informed consent.
Collapse
Affiliation(s)
- Holly L Peay
- RTI International, 3040 E Cornwallis Rd., Research Triangle Park, Durham, NC, 27709, USA.
| | - Stuart Rennie
- Social Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R Jean Cadigan
- Social Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angela Gwaltney
- RTI International, 3040 E Cornwallis Rd., Research Triangle Park, Durham, NC, 27709, USA
| | - Thidarat Jupimai
- Center of Excellence in Pediatric Infectious Diseases and Vaccines Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nittaya Phanuphak
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Eugène Kroon
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Donn J Colby
- Institute of HIV Research and Innovation, Bangkok, Thailand
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Nuchanart Ormsby
- Social Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sinéad C Isaacson
- Social Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sandhya Vasan
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Carlo Sacdalan
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Peeriya Prueksakaew
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Khunthalee Benjapornpong
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Jintanat Ananworanich
- Department of Global Health, Amsterdam University Medical Centers, University of Amsterdam, and Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Gail E Henderson
- Social Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
13
|
Dashti A, Singh V, Chahroudi A. HIV Reservoirs: Modeling, Quantification, and Approaches to a Cure. Methods Mol Biol 2022; 2407:215-228. [PMID: 34985668 DOI: 10.1007/978-1-0716-1871-4_16] [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: 06/14/2023]
Abstract
Biomedical research in animal models depends heavily on nonhuman primates (NHP) (Phillips et al., Am J Primatol 76(9):801-827, 2014). In their physiology, neurobiology, and, most importantly, their susceptibility to infectious diseases and subsequent immune responses, NHPs have many parallels with humans (Rhesus Macaque Genome Sequencing and Analysis Consortium et al., Science 316(5822):222-234, 2007). Different species of NHPs have served as important animal models for numerous infectious diseases spanning a wide range of pathogens (Gardner and Luciw, ILAR J 49(2):220-255, 2008). As a result of recognizing their utility in HIV research, NHPs have contributed to groundbreaking studies of disease pathogenesis, vaccination, and curative research (London et al., Lancet 2(8355):869-873, 1983; Henrickson et al., Lancet 1 (8321):388-390, 1983). Many African NHPs are considered natural hosts for SIV in which SIV infection is usually nonprogressive and does not cause acquired immunodeficiency syndrome (AIDS) (Chahroudi et al., Science 335(6073):1188-1193, 2012; Taaffe et al., J Virol 84(11):5476-5484, 2010). However, cross-species transmission of SIV strains to other NHPs or to humans (nonnatural hosts) leads to progressive disease and AIDS (Paiardini et al., Annu Rev Med 60:485-495, 2009). In particular, SIV infection of Asian rhesus macaques recapitulates many features of HIV infection in humans and therefore has become a widely used approach for contemporary HIV research into virus persistence and cure strategies (Gardner and Luciw, FASEB J 3(14):2593-2606, 1989). There are multiple factors that should be considered in HIV/SIV studies using NHPs including the particular monkey species and geographic background, age and sex, certain genetic properties, virus strain, route and dose of infection, interventional treatments, and prespecified study outcomes. Here, we discuss consideration of these factors to address specific questions in HIV cure research.
Collapse
Affiliation(s)
- Amir Dashti
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Vidisha Singh
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Ann Chahroudi
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
New Approaches to Multi-Parametric HIV-1 Genetics Using Multiple Displacement Amplification: Determining the What, How, and Where of the HIV-1 Reservoir. Viruses 2021; 13:v13122475. [PMID: 34960744 PMCID: PMC8709494 DOI: 10.3390/v13122475] [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: 10/29/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/27/2022] Open
Abstract
Development of potential HIV-1 curative interventions requires accurate characterization of the proviral reservoir, defined as host-integrated viral DNA genomes that drive rebound of viremia upon halting ART (antiretroviral therapy). Evaluation of such interventions necessitates methods capable of pinpointing the rare, genetically intact, replication-competent proviruses within a background of defective proviruses. This evaluation can be achieved by identifying the distinct integration sites of intact proviruses within host genomes and monitoring the dynamics of these proviruses and host cell lineages over longitudinal sampling. Until recently, molecular genetic approaches at the single proviral level have been generally limited to one of a few metrics, such as proviral genome sequence/intactness, host-proviral integration site, or replication competency. New approaches, taking advantage of MDA (multiple displacement amplification) for WGA (whole genome amplification), have enabled multiparametric proviral characterization at the single-genome level, including proviral genome sequence, host-proviral integration site, and phenotypic characterization of the host cell lineage, such as CD4 memory subset and antigen specificity. In this review, we will examine the workflow of MDA-augmented molecular genetic approaches to study the HIV-1 reservoir, highlighting technical advantages and flexibility. We focus on a collection of recent studies in which investigators have used these approaches to comprehensively characterize intact and defective proviruses from donors on ART, investigate mechanisms of elite control, and define cell lineage identity and antigen specificity of infected CD4+ T cell clones. The highlighted studies exemplify how these approaches and their future iterations will be key in defining the targets and evaluating the impacts of HIV curative interventions.
Collapse
|
15
|
Siliciano JD, Siliciano RF. In Vivo Dynamics of the Latent Reservoir for HIV-1: New Insights and Implications for Cure. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:271-294. [PMID: 34736342 DOI: 10.1146/annurev-pathol-050520-112001] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although antiretroviral therapy (ART) can reduce viremia to below the limit of detection and allow persons living with HIV-1 (PLWH) to lead relatively normal lives, viremia rebounds when treatment is interrupted. Rebound reflects viral persistence in a stable latent reservoir in resting CD4+ T cells. This reservoir is now recognized as the major barrier to cure and is the focus of intense international research efforts. Strategies to cure HIV-1 infection include interventions to eliminate this reservoir, to prevent viral rebound from the reservoir, or to enhance immune responses such that viral replication is effectively controlled. Here we consider recent developments in understanding the composition of the reservoir and how it can be measured in clinical studies. We also discuss exciting new insights into the in vivo dynamics of the reservoir and the reasons for its remarkable stability. Finally we discuss recent discoveries on the complex processes that govern viral rebound. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Janet D Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; .,Howard Hughes Medical Institute, Baltimore, Maryland 21205, USA
| |
Collapse
|
16
|
Gao P, Yu F, Yang X, Li D, Shi Y, Wang Y, Zhang F. Evaluation of a novel in-house HIV-1 genotype drug resistance assay using clinical samples in China. Curr HIV Res 2021; 20:32-41. [PMID: 34515004 PMCID: PMC9127726 DOI: 10.2174/1570162x19666210910144433] [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: 01/30/2021] [Revised: 07/08/2021] [Accepted: 08/04/2021] [Indexed: 11/22/2022]
Abstract
Background HIV drug resistance poses a major challenge for anti-retroviral treatment (ART) and the prevention and control of HIV epidemic. Objective The study aims to establish a novel in-house assay with high efficiency, named AP in- house method, that would be suitable for HIV-1 drug resistance detection in China. Methods An in-house HIV-1 genotyping method was used to sequence the partial pol gene from 60 clinical plasma samples; the results of our test were compared with a commercial ViroSeq HIV-1 genotyping system. Results Among sixty samples, 58(96.7%) were successfully amplified by AP in-house method, five of them harbored viral load below 1,000 copies/ml. The genotype distribution was 43.1% CRF07_BC (25/58), 39.7% CRF01_AE (23/58), 6.9% CRF55_01B (4/58), 5.2% subtype B (3/58) and 5.2% CRF08_BC (3/58). Compared with that of the ViroSeq system, the consistent rate of these nucleotides and amino acids obtained by AP in-house method was up to 99.5 ± 0.4% and 99.5 ± 0.4%, respectively. A total of 290 HIV-1 drug resistance mutations were identified by two methods, including 126 nucleoside reverse transcriptase inhibitors (NRTIs), 145 non-nucleoside reverse transcriptase inhibitors (NNRTIs) and 19 protease inhibitors (PIs) resistance mutations. Out of them, 94.1% (273/290) were completely concordant between the AP in-house method and the ViroSeq system. Conclusion Overall, the evaluation of AP in-house method provided comparable results to those of the ViroSeq system on diversified HIV-1 subtypes in China.
Collapse
Affiliation(s)
- Peijie Gao
- Beijing Anapure Bioscitific Co. Ltd. Beijing. China
| | - Fengting Yu
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital. China
| | | | - Dan Li
- Beijing Anapure Bioscitific Co. Ltd. Beijing. China
| | - Yalun Shi
- Beijing Anapure Bioscitific Co. Ltd. Beijing. China
| | - Yan Wang
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital. China
| | - Fujie Zhang
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital. China
| |
Collapse
|
17
|
Analytical Treatment Interruption in HIV Trials: Statistical and Study Design Considerations. Curr HIV/AIDS Rep 2021; 18:475-482. [PMID: 34213731 PMCID: PMC8251690 DOI: 10.1007/s11904-021-00569-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
Purpose of Review Analytical treatment interruption (ATI) remains an essential component in clinical studies investigating novel agents or combination treatment strategies aiming to induce HIV treatment-free remission or long-term viral control. We provide an overview on key study design aspects of ATI trials from the perspective of statisticians. Recent Findings ATI trial designs have evolved towards shorter treatment interruption phases and more frequent viral load monitoring aiming to reduce prolonged viremia risks. Criteria for ART resumption have evolved as well. Common outcome measures in modern ATI trials include time to viral rebound, viral control, and viral set point. Summary Design of the ATI component in HIV clinical trials is driven by the scientific question and the mechanism of action of the intervention being investigated.
Collapse
|
18
|
Diepstra KL, Barr L, Palm D, Hogg E, Mollan KR, Henley L, Stover AM, Simoni JM, Sugarman J, Brown B, Sauceda JA, Deeks S, Fox L, Gandhi RT, Smith D, Li JZ, Dubé K. Participant Perspectives and Experiences Entering an Intensively Monitored Antiretroviral Pause: Results from the AIDS Clinical Trials Group A5345 Biomarker Study. AIDS Res Hum Retroviruses 2021; 37:489-501. [PMID: 33472545 DOI: 10.1089/aid.2020.0222] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The AIDS Clinical Trials Group (ACTG) A5345 study included an intensively monitored antiretroviral pause (IMAP), during which a cohort of participants temporarily stopped antiretroviral treatment during chronic HIV infection. We surveyed participant perceptions and understanding of A5345 using a cross-sectional sociobehavioral questionnaire. Participants completed the baseline questionnaire either before or after initiating the study's IMAP. Questionnaire responses were linked to existing demographic data. Quantitative responses were analyzed overall and stratified by IMAP status. Open-ended responses were analyzed using conventional content analysis. Thirty-two participants completed the baseline sociobehavioral questionnaire. Half (n = 16) completed it before (i.e., pre-IMAP initiation group) and half (n = 16) after IMAP initiation (i.e., post-IMAP initiation group). Eight pre-IMAP initiation respondents (50%) and 11 post-IMAP respondents (69%) responded "yes" when asked if they perceived any direct benefits from participating in A5345. Perceived societal-level benefits included furthering HIV cure-related research and helping the HIV community. Perceived personal-level benefits included the opportunity to learn about the body's response to IMAP and financial compensation. The majority of respondents-13 from each group (81% of each)-reported risks from participation, for example, viral load becoming detectable. A5345 participants perceived both societal- and personal-level benefits of study participation. While the majority of survey respondents perceived participatory risks, nearly one in five did not. Key messages pertaining to study-related risks and benefits may need to be clarified or reiterated periodically throughout follow-up in HIV cure-related studies with IMAPs. Clinical Trail Registration Number: NCT03001128.
Collapse
Affiliation(s)
- Karen L. Diepstra
- UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Liz Barr
- AIDS Clinical Trials Group (ACTG) Community Scientific Sub-Committee, Baltimore, Maryland, USA
| | - David Palm
- Institute of Global Health and Infectious Diseases (IGHID), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Evelyn Hogg
- Social & Scientific Systems, a DLH Company, Silver Spring, Maryland, USA
| | - Katie R. Mollan
- Center for AIDS Research (CFAR), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Laney Henley
- UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Angela M. Stover
- UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | - Jane M. Simoni
- Department of Psychology, University of Washington, Seattle, Washington, USA
- Department of Global Health and Gender, Women, and Sexuality Studies, University of Washington, Seattle, Washington, USA
| | - Jeremy Sugarman
- Johns Hopkins Berman Institute for Bioethics, Baltimore, Maryland, USA
| | - Brandon Brown
- Department of Social Medicine, Population and Public Health, Center for Healthy Communities, University of California, Riverside School of Medicine, Riverside, California, USA
| | - John A. Sauceda
- Division of Prevention Sciences, Center for AIDS Prevention Studies (CAPS), University of California, San Francisco, San Francisco, California, USA
| | - Steven Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Lawrence Fox
- National Institute of Health (NIH) Division of AIDS (DAIDS), Bethesda, Maryland, USA
| | | | - Davey Smith
- Division of Infectious Diseases and Global Health, University of California, San Diego, California, USA
| | - Jonathan Z. Li
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Karine Dubé
- UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| |
Collapse
|
19
|
Immonen TT, Fennessey CM, Lipkey L, Thorpe A, Del Prete GQ, Lifson JD, Davenport MP, Keele BF. Transient viral replication during analytical treatment interruptions in SIV infected macaques can alter the rebound-competent viral reservoir. PLoS Pathog 2021; 17:e1009686. [PMID: 34143853 PMCID: PMC8244872 DOI: 10.1371/journal.ppat.1009686] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/30/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Analytical treatment interruptions (ATIs) of antiretroviral therapy (ART) play a central role in evaluating the efficacy of HIV-1 treatment strategies targeting virus that persists despite ART. However, it remains unclear if ATIs alter the rebound-competent viral reservoir (RCVR), the virus population that persists during ART and from which viral recrudescence originates after ART discontinuation. To assess the impact of ATIs on the RCVR, we used a barcode sequence tagged SIV to track individual viral lineages through a series of ATIs in Rhesus macaques. We demonstrate that transient replication of individual rebounding lineages during an ATI can lead to their enrichment in the RCVR, increasing their probability of reactivating again after treatment discontinuation. These data establish that the RCVR can be altered by uncontrolled replication during ATI.
Collapse
Affiliation(s)
- Taina T. Immonen
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Christine M. Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Leslie Lipkey
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Abigail Thorpe
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Gregory Q. Del Prete
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Miles P. Davenport
- Infection Analytics Program, Kirby Institute for Infection and Immunity, University of New South Wales, Sydney, New South Wales, Australia
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| |
Collapse
|
20
|
Henderson GE, Rennie S, Corneli A, Peay HL. Cohorts as collections of bodies and communities of persons: insights from the SEARCH010/RV254 research cohort. Int Health 2021; 12:584-590. [PMID: 33165552 PMCID: PMC7650957 DOI: 10.1093/inthealth/ihaa060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/28/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Longitudinal research cohorts are uniquely suited to answer research questions about morbidity and mortality. Cohorts may be comprised of individuals identified by specific conditions or other shared traits. We argue that research cohorts are more than simply aggregations of individuals and their associated data to meet research objectives. They are social communities comprised of members, investigators and organizations whose own interests, identities and cultures interact and evolve over time. The literature describes a range of scientific and ethical challenges and opportunities associated with cohorts. To advance these deliberations, we report examples from the literature and our own research on the Thai SEARCH010/RV254 cohort, comprising individuals diagnosed with human immunodeficiency virus (HIV) during acute infection. We reflect on the impact of cohort experiences and identity, and specifically how people incorporate cohort participation into meaning making associated with their diagnosis, the influence of cohort participation on decision making for early-phase clinical trials recruited from within the cohort, and the impact of the relationships that exist between researchers and participants. These data support the concept of cohorts as communities of persons, where identity is shaped, in part, through cohort experiences. The social meanings associated with cohorts have implications for the ethics of cohort-based research, as social contexts inevitably affect the ways that ethical concerns manifest.
Collapse
Affiliation(s)
- Gail E Henderson
- Department of Social Medicine, University of North Carolina School of Medicine, 347A MacNider, 333 South Columbia Street, Chapel Hill, NC 27599-7240, USA
| | - Stuart Rennie
- Department of Social Medicine, University of North Carolina School of Medicine, 347A MacNider, 333 South Columbia Street, Chapel Hill, NC 27599-7240, USA
| | - Amy Corneli
- Departments of Population Health Sciences and Medicine, School of Medicine, Duke Clinical Research Institute, Duke University, 215 Morris Street, Durham, NC 27701
| | - Holly L Peay
- RTI International, 3040 E Cornwallis Rd, Research Triangle Park, NC 27709
| |
Collapse
|
21
|
Peay HL, Ormsby NQ, Henderson GE, Jupimai T, Rennie S, Siripassorn K, Kanchawee K, Isaacson S, Cadigan RJ, Kuczynski K, Likhitwonnawut U. Recommendations from Thai stakeholders about protecting HIV remission ('cure') trial participants: report from a participatory workshop. Int Health 2021; 12:567-574. [PMID: 33165551 PMCID: PMC7650909 DOI: 10.1093/inthealth/ihaa067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Accepted: 09/02/2020] [Indexed: 12/02/2022] Open
Abstract
Background The social/behavioral HIV Decision-Making Study (DMS) assesses informed consent and trial experiences of individuals in HIV remission trials in Thailand. We convened a 1-d multi-stakeholder participatory workshop in Bangkok. We provide a meeting summary and reactions from DMS investigators. Methods Workshop members viewed de-identified interview excerpts from DMS participants. They deliberated on the findings and made recommendations regarding informed choice for remission trials. Notes and recordings were used to create a summary report, which was reviewed by members and refined. Results Workshop members’ recommendations included HIV education and psychosocial support to establish the basis for informed choice, key trial information to be provided in everyday language, supportive decision-making processes and psychosocial care during and after the trial. Concerns included participant willingness to restart antiretrovirals after trial-mandated treatment interruption, unintended influence of the research team on decision-making and seemingly altruistic motivations for trial participation that may signal attempts to atone for stigmatized behavior. Conclusions The workshop highlighted community perspectives and resulted in recommendations for supporting informed choice and psychosocial and physical health. These are the first such recommendations arising from a deliberative process. Although some elements are rooted in the Thai context, most are applicable across remission trials.
Collapse
Affiliation(s)
| | - Nuchanart Q Ormsby
- University of North Carolina at Chapel Hill, Department of Social Medicine, USA
| | - Gail E Henderson
- University of North Carolina at Chapel Hill, Department of Social Medicine, USA
| | - Thidarat Jupimai
- Center of Excellence in Pediatric Infectious Diseases and Vaccines Faculty of Medicine, Chulalongkorn University, Thailand
| | - Stuart Rennie
- University of North Carolina at Chapel Hill, Department of Social Medicine, USA.,University of North Carolina at Chapel Hill, Center for Bioethics, USA
| | | | - Kunakorn Kanchawee
- Center of Excellence in Research on Gender, Sexuality and Health, Faculty of Social Sciences and Humanities, Mahidol University, Thailand
| | - Sinéad Isaacson
- University of North Carolina at Chapel Hill, Department of Social Medicine, USA.,University of North Carolina at Chapel Hill, Department of Epidemiology, USA
| | - R Jean Cadigan
- University of North Carolina at Chapel Hill, Department of Social Medicine, USA.,University of North Carolina at Chapel Hill, Center for Bioethics, USA
| | - Kriste Kuczynski
- University of North Carolina at Chapel Hill, Department of Social Medicine, USA
| | | |
Collapse
|
22
|
Nsoh M, Tshimwanga KE, Ngum BA, Mgasa A, Otieno MO, Moali B, Sirili N, Atanga NS, Halle-Ekane GE. Predictors of antiretroviral therapy interruptions and factors influencing return to care at the Nkolndongo Health District, Cameroon. Afr Health Sci 2021; 21:29-38. [PMID: 34447421 PMCID: PMC8367305 DOI: 10.4314/ahs.v21i1.6s] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Antiretroviral therapy is a lifelong commitment that requires consistent intake of tablets to optimize health outcomes, attain and maintain viral suppression. OBJECTIVE We aimed to elicit predictors of treatment interruption amongst PLHIV and identify motivating factors influencing return to care. METHOD We conducted a cross-sectional study using a mixed-method approach in four hospitals in Yaoundé. Sociodemographic and clinical data were collected from ART registers. Using purposeful sampling, thirteen participants were enrolled for interviews. Quantitative data were analyzed using Epi-Info and Atlas-TI for qualitative analysis. Ethical clearance approved by CBCHS-IRB. RESULTS A total of 271 participants records were assessed. The mean age was 33 years (SD±11years). Private facilities CASS and CMNB registered respectively 53 (19.6%) and 14 (5.2%) participants while CMA Nkomo and IPC had 114 (42.1%) and 90 (33.2%) participants. Most participants (75.3%) were females [OR 1.14; CI 0.78-1.66] compare with males. 78% had no viral load test results. Transport cost and stigmatization constituted the most prominent predictors of treatment interruption (47.5%) and (10.5%) respectively. Belief in the discovery of an eminent HIV cure and the desire to raise offspring motivated 30% and 61%, respectively to resume treatment. CONCLUSION Structural barriers like exposed health facility, and dispensing ARVs in open spaces stigmatizes clients and increases odds of attrition. Attrition of patients on ART will be minimized through implementation of client centered approaches like multiplying proxy ART pick points, devolving stable clients to community ARV model.
Collapse
Affiliation(s)
- Marius Nsoh
- Department of Public Health, School of Health Sciences, Catholic University of Central Africa; Cameroon
- HIV Free Project, Cameroon Baptist Convention Health Services, Center region; Cameroon
| | - Katayi E Tshimwanga
- HIV Free Project, Cameroon Baptist Convention Health Services, Center region; Cameroon
| | - Busi A Ngum
- Women Health Program, Mbingo Baptist Hospital, Cameroon Baptist Convention Health Services; Cameroon
| | - Avelina Mgasa
- Ministry of Health Community Development, Gender, Elderly and Children; National Blood Transfusion Service; Tanzania
| | - Moses O Otieno
- National AIDS and Sexually Transmitted Infections Control Program (NASCOP); Kenya
| | - Bokwena Moali
- Ministry of Health and Wellness, Okavango District, Botswana
| | - Nathanael Sirili
- Department of Development Studies, Muhimbili University of Health and Allied Sciences; Tanzania
| | - Ndeso S Atanga
- Department of Public Health, Obstetrics and Gynecology, Faculty of Health Sciences, University of Buea; Cameroon
| | - Gregory E Halle-Ekane
- Department of Public Health, Obstetrics and Gynecology, Faculty of Health Sciences, University of Buea; Cameroon
| |
Collapse
|
23
|
Increased Proviral DNA in Circulating Cells Correlates with Plasma Viral Rebound in Simian Immunodeficiency Virus-Infected Rhesus Macaques after Antiretroviral Therapy Interruption. J Virol 2021; 95:JVI.02064-20. [PMID: 33408173 PMCID: PMC8094949 DOI: 10.1128/jvi.02064-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/18/2020] [Indexed: 12/30/2022] Open
Abstract
Viral reservoirs are involved in persistent HIV infection, and a small number of mosaic latent cellular reservoirs promote viral rebound upon analytical treatment interruption, which is the major obstacle to a cure. However, early indicators that can predict resurgence of viremia after treatment interruption may aid treatment decisions in people living with HIV. The human immunodeficiency virus (HIV) reservoir is responsible for persistent viral infection, and a small number of mosaic latent cellular reservoirs promote viral rebound upon antiretroviral therapy interruption, which is the major obstacle to a cure. However, markers that determine effective therapy and viral rebound posttreatment interruption remain unclear. In this study, we comprehensively and longitudinally tracked dynamic decay of cell-associated viral RNA/DNA in systemic and lymphoid tissues in simian immunodeficiency virus (SIV)-infected rhesus macaques on prolonged combined antiretroviral therapy (cART) and evaluated predictors of viral rebound after treatment cessation. The results showed that suppressive ART substantially reduced plasma SIV RNA, cell-associated unspliced, and multiply spliced SIV RNA to undetectable levels, yet viral DNA remained detectable in systemic tissues and lymphoid compartments throughout cART. Intriguingly, a rapid increase of integrated proviral DNA in peripheral mononuclear cells was detected once treatment was withdrawn, accompanied by the emergence of detectable plasma viral load. Notably, the increase of peripheral proviral DNA after treatment interruption correlated with the emergence and degree of viral rebound. These findings suggest that measuring total viral DNA in SIV infection may be a relatively simple surrogate marker of reservoir size and may predict viral rebound after treatment interruption and inform treatment strategies. IMPORTANCE Viral reservoirs are involved in persistent HIV infection, and a small number of mosaic latent cellular reservoirs promote viral rebound upon analytical treatment interruption, which is the major obstacle to a cure. However, early indicators that can predict resurgence of viremia after treatment interruption may aid treatment decisions in people living with HIV. Utilizing the rhesus macaque model, we demonstrated that increased proviral DNA in peripheral cells after treatment interruption, rather than levels of proviral DNA, was a useful marker to predict the emergence and degree of viral rebound after treatment interruption, providing a rapid approach for monitoring HIV rebound and informing decisions.
Collapse
|
24
|
Prolonged administration of maraviroc reactivates latent HIV in vivo but it does not prevent antiretroviral-free viral rebound. Sci Rep 2020; 10:22286. [PMID: 33339855 PMCID: PMC7749169 DOI: 10.1038/s41598-020-79002-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/27/2020] [Indexed: 01/24/2023] Open
Abstract
Human immunodeficiency virus (HIV) remains incurable due to latent viral reservoirs established in non-activated CD4 T cells that cannot be eliminated via antiretroviral therapy. Current efforts to cure HIV are focused on identifying drugs that will induce viral gene expression in latently infected cells, commonly known as latency reversing agents (LRAs). Some drugs have been shown to reactivate latent HIV but do not cause a reduction in reservoir size. Therefore, finding new LRAs or new combinations or increasing the round of stimulations is needed to cure HIV. However, the effects of these drugs on viral rebound after prolonged treatment have not been evaluated. In a previous clinical trial, antiretroviral therapy intensification with maraviroc for 48 weeks caused an increase in residual viremia and episomal two LTR-DNA circles suggesting that maraviroc could reactivate latent HIV. We amended the initial clinical trial to explore additional virologic parameters in stored samples and to evaluate the time to viral rebound during analytical treatment interruption in three patients. Maraviroc induced an increase in cell-associated HIV RNA during the administration of the drug. However, there was a rapid rebound of viremia after antiretroviral therapy discontinuation. HIV-specific T cell response was slightly enhanced. These results show that maraviroc can reactivate latent HIV in vivo but further studies are required to efficiently reduce the reservoir size.
Collapse
|
25
|
Saha A, Dixit NM. Pre-existing resistance in the latent reservoir can compromise VRC01 therapy during chronic HIV-1 infection. PLoS Comput Biol 2020; 16:e1008434. [PMID: 33253162 PMCID: PMC7728175 DOI: 10.1371/journal.pcbi.1008434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 12/10/2020] [Accepted: 10/11/2020] [Indexed: 01/26/2023] Open
Abstract
Passive immunization with broadly neutralizing antibodies (bNAbs) of HIV-1 appears a promising strategy for eliciting long-term HIV-1 remission. When administered concomitantly with the cessation of antiretroviral therapy (ART) to patients with established viremic control, bNAb therapy is expected to prolong remission. Surprisingly, in clinical trials on chronic HIV-1 patients, the bNAb VRC01 failed to prolong remission substantially. Identifying the cause of this failure is important for improving VRC01-based therapies and unraveling potential vulnerabilities of other bNAbs. In the trials, viremia resurged rapidly in most patients despite suppressive VRC01 concentrations in circulation, suggesting that VRC01 resistance was the likely cause of failure. ART swiftly halts viral replication, precluding the development of resistance during ART. If resistance were to emerge post ART, virological breakthrough would have taken longer than without VRC01 therapy. We hypothesized therefore that VRC01-resistant strains must have been formed before ART initiation, survived ART in latently infected cells, and been activated during VRC01 therapy, causing treatment failure. Current assays preclude testing this hypothesis experimentally. We developed a mathematical model based on the hypothesis and challenged it with available clinical data. The model integrated within-host HIV-1 evolution, stochastic latency reactivation, and viral dynamics with multiple-dose VRC01 pharmacokinetics. The model predicted that single but not higher VRC01-resistant mutants would pre-exist in the latent reservoir. We constructed a virtual patient population that parsimoniously recapitulated inter-patient variations. Model predictions with this population quantitatively captured data of VRC01 failure from clinical trials, presenting strong evidence supporting the hypothesis. We attributed VRC01 failure to single-mutant VRC01-resistant proviruses in the latent reservoir triggering viral recrudescence, particularly when VRC01 was at trough levels. Pre-existing resistant proviruses in the latent reservoir may similarly compromise other bNAbs. Our study provides a framework for designing bNAb-based therapeutic protocols that would avert such failure and maximize HIV-1 remission. Antiretroviral therapy (ART) can control but not eradicate HIV-1. Stopping ART leads to rapid viral resurgence and progressive disease. ART is therefore administered lifelong. Tremendous efforts are ongoing to devise strategies that will enable stopping ART and yet prevent viral resurgence. One such strategy involves the administration of broadly neutralizing antibodies (bNAbs) of HIV-1 at the time of stopping ART. This strategy is expected to delay if not prevent viral resurgence. Surprisingly, treatment with VRC01, a potent bNAb, resulted in hardly any improvement in viral remission. In this study, we elucidate the cause of this failure. We hypothesized that VRC01-resistant strains may pre-exist in latently infected cells, which are unaffected by ART. They can thus outlast ART and get reactivated, triggering VRC01 failure. We built a detailed mathematical model based on this hypothesis and showed that it quantitatively captured observations of VRC01 failure in clinical trials on chronic HIV-1 patients. Our study thus identifies a potential vulnerability of bNAbs, namely, bNAb-resistant strains pre-existing in latently infected cells. Our model offers a framework for predicting bNAb-based treatment protocols that would preclude failure due to pre-existing resistance and maximally prolong remission.
Collapse
Affiliation(s)
- Ananya Saha
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| | - Narendra M. Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, India
- * E-mail:
| |
Collapse
|
26
|
Wilson A, Lynch RM. Embracing diversity: how can broadly neutralizing antibodies effectively target a diverse HIV-1 reservoir? Curr Opin Pharmacol 2020; 54:173-178. [PMID: 33189993 DOI: 10.1016/j.coph.2020.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
Genetic diversity in the latent proviral reservoir of HIV-1 infected individuals poses a challenge to cure strategies. It has become increasingly evident that diversity increases proportionally with length of active infection, and that functional and/or sterilizing cure strategies will need to overcome this obstacle in individuals who initiated antiretroviral therapy (ART) during chronic infection. Analyzing the results of analytic treatment interruption (ATI) has allowed for the evaluation of such therapeutic strategies in HIV+ individuals. Strategies to overcome the genetic diversity of the HIV-1 reservoir include antibody combinations, pre-screening individuals for bNAb sensitivity, focusing on low-diversity individuals as well as targeting host proteins.
Collapse
Affiliation(s)
- Andrew Wilson
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine & Health Sciences, Washington DC, USA
| | - Rebecca M Lynch
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine & Health Sciences, Washington DC, USA.
| |
Collapse
|
27
|
Leal L, Fehér C, Richart V, Torres B, García F. Antiretroviral Therapy Interruption (ATI) in HIV-1 Infected Patients Participating in Therapeutic Vaccine Trials: Surrogate Markers of Virological Response. Vaccines (Basel) 2020; 8:vaccines8030442. [PMID: 32764508 PMCID: PMC7564579 DOI: 10.3390/vaccines8030442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022] Open
Abstract
A functional Human immunodeficiency Virus (HIV) cure has been proposed as an alternative to antiretroviral treatment for life, and therapeutic vaccines represent one of the most promising approaches. The goal of therapeutic vaccination is to augment virus-specific immune responses that have an impact on HIV viral load dynamics. To date, the agreed feature to evaluate the effects of these therapeutic interventions is analytical antiretroviral treatment interruption (ATI), at least until we find a reliable biomarker that can predict viral control. Different host, immunologic, and virologic markers have been proposed as predictors of viral control during ATI after therapeutic interventions. This review describes the relevance of ATI and the different surrogate markers of virological control assessed in HIV therapeutic vaccine clinical trials.
Collapse
Affiliation(s)
- Lorna Leal
- Infectious Diseases Department—HIV Unit, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (C.F.); (V.R.); (B.T.); (F.G.)
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-2275586; Fax: +34-93-4514-438
| | - Csaba Fehér
- Infectious Diseases Department—HIV Unit, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (C.F.); (V.R.); (B.T.); (F.G.)
| | - Valèria Richart
- Infectious Diseases Department—HIV Unit, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (C.F.); (V.R.); (B.T.); (F.G.)
| | - Berta Torres
- Infectious Diseases Department—HIV Unit, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (C.F.); (V.R.); (B.T.); (F.G.)
| | - Felipe García
- Infectious Diseases Department—HIV Unit, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (C.F.); (V.R.); (B.T.); (F.G.)
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
28
|
Abstract
Therapeutic approaches towards a functional cure or eradication of HIV have gained renewed momentum upon encouraging data emerging from studies in SIV monkey models and recent results from human clinical studies. However, a multitude of questions remain to be addressed, including how to deal with the latent viral reservoir, how to boost the host immune response to the virus and what the hurdles are to reach relevant viral compartments in the body. Advances have been made especially with regard to identifying agents that can reactivate the latent virus in vivo and boost the cellular and humoral immunity, but it remains largely unclear whether any of these strategies can awaken a sufficiently large fraction of the viral reservoir and whether the boosted immunity can prevent rapid viral replication once antiretroviral treatments are stopped.
Collapse
Affiliation(s)
- Lucia Bailon
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Beatriz Mothe
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain
| | | | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.
- Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.
- AELIX Therapeutics, Barcelona, Spain.
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
29
|
Lelièvre JD. Preexposure Prophylaxis for Mitigating Risk of HIV Transmission During HIV Cure-Related Clinical Trials With a Treatment Interruption. J Infect Dis 2020; 220:S16-S18. [PMID: 30860581 DOI: 10.1093/infdis/jiz036] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 02/04/2019] [Indexed: 01/18/2023] Open
Abstract
Analytical treatment interruption performed during human immunodeficiency virus (HIV) cure-related clinical trials exposes sex partners of participants in these trials to a risk of HIV transmission. Preexposure prophylaxis (PrEP), which emerged in recent years as a key strategy for preventing HIV transmission, is often considered a useful tool to prevent this risk. This article supports offering PrEP to the stable sex partners of participants in these trials but also notes limitations that must be addressed. It concludes that PrEP cannot on its own eliminate the risk of secondary transmission in this context.
Collapse
Affiliation(s)
- Jean-Daniel Lelièvre
- Vaccine Research Institute, Université Paris Est Créteil.,INSERM U955, équipe 16, Faculté de Médecine, Université Paris Est Créteil.,Assistance Publique-Hôpitaux de Paris, Service d'Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Créteil, France
| |
Collapse
|
30
|
Di Sante L, Costantini A, Caucci S, Corsi A, Brescini L, Menzo S, Bagnarelli P. Quantification of the HIV-1 total reservoir in the peripheral blood of naïve and treated patients by a standardised method derived from a commercial HIV-1 RNA quantification assay. Clin Chem Lab Med 2020; 59:609-617. [PMID: 33326413 DOI: 10.1515/cclm-2020-0142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/05/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES HIV-1 DNA can persist in host cells, establishing a latent reservoir. This study was aimed to develop an extraction and amplification protocol for HIV-1 DNA quantification by modifying a quantitative commercial assay. METHODS HIV-1 DNA was extracted on an Abbott m2000sp instrument, using an open-mode protocol. Two calibrators, spiked with a plasmid containing HIV-1 genome (103 and 105 cps/mL), were extracted and amplified to generate a master calibration curve. Precision, accuracy, linear dynamic range, limit of quantification (LOQ) and limit of detection (LOD) were determined. A cohort of patients, naïve or chronically infected, was analysed. RESULTS Calibration curve was obtained from 42 replicates of standards (stds); precision was calculated (coefficients of variability [CVs] below 10%); accuracy was higher than 90%. Linearity covered the entire range tested (10-104 copies per reaction), and LOD (95%) was 12 copies per reaction. HIV-1 DNA was significantly higher (p < 0.0001) in drug-naïve (62) than in chronically treated patients (50), and proviral loads correlated with lymphocytes (p = 0.0002) and CD4+ (p < 0.0001) counts only in naïve patients. Both groups displayed a significant inverse correlation between CD4+ nadir and proviral loads. A significant correlation (p < 0.0001) between viraemia and HIV-1 reservoir was disclosed. No significant difference was obtained from the comparison between proviral loads on whole blood and peripheral blood mononuclear cells (PBMCs) from the same patient. CONCLUSIONS The novelty of our approach relies on the selection of appropriate reference standard extracted and amplified as clinical specimens avoiding any underestimation of the reservoir. Results confirm HIV-1 DNA as a marker of disease progression, supporting the relationship between the width of latent reservoir and the immunological status of the patient.
Collapse
Affiliation(s)
- Laura Di Sante
- Department of Molecular and Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Andrea Costantini
- Department of Molecular and Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Sara Caucci
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Alice Corsi
- Department of Molecular and Clinical Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Lucia Brescini
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Stefano Menzo
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Patrizia Bagnarelli
- Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
31
|
Fidler S, Stöhr W, Pace M, Dorrell L, Lever A, Pett S, Kinloch-de Loes S, Fox J, Clarke A, Nelson M, Thornhill J, Khan M, Fun A, Bandara M, Kelly D, Kopycinski J, Hanke T, Yang H, Bennett R, Johnson M, Howell B, Barnard R, Wu G, Kaye S, Wills M, Babiker A, Frater J. Antiretroviral therapy alone versus antiretroviral therapy with a kick and kill approach, on measures of the HIV reservoir in participants with recent HIV infection (the RIVER trial): a phase 2, randomised trial. Lancet 2020; 395:888-898. [PMID: 32085823 DOI: 10.1016/s0140-6736(19)32990-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/25/2019] [Accepted: 11/15/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Antiretroviral therapy (ART) cannot cure HIV infection because of a persistent reservoir of latently infected cells. Approaches that force HIV transcription from these cells, making them susceptible to killing-termed kick and kill regimens-have been explored as a strategy towards an HIV cure. RIVER is the first randomised trial to determine the effect of ART-only versus ART plus kick and kill on markers of the HIV reservoir. METHODS This phase 2, open-label, multicentre, randomised, controlled trial was undertaken at six clinical sites in the UK. Patients aged 18-60 years who were confirmed as HIV-positive within a maximum of the past 6 months and started ART within 1 month from confirmed diagnosis were randomly assigned by a computer generated randomisation list to receive ART-only (control) or ART plus the histone deacetylase inhibitor vorinostat (the kick) and replication-deficient viral vector T-cell inducing vaccines encoding conserved HIV sequences ChAdV63. HIVconsv-prime and MVA.HIVconsv-boost (the kill; ART + V + V; intervention). The primary endpoint was total HIV DNA isolated from peripheral blood CD4+ T-cells at weeks 16 and 18 after randomisation. Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, NCT02336074. FINDINGS Between June 14, 2015 and Jul 11, 2017, 60 men with HIV were randomly assigned to receive either an ART-only (n=30) or an ART + V + V (n=30) regimen; all 60 participants completed the study, with no loss-to-follow-up. Mean total HIV DNA at weeks 16 and 18 after randomisation was 3·02 log10 copies HIV DNA per 106 CD4+ T-cells in the ART-only group versus 3·06 log10 copies HIV DNA per 106 CD4+ T-cells in ART + V + V group, with no statistically significant difference between the two groups (mean difference of 0·04 log10 copies HIV DNA per 106 CD4+ T-cells [95% CI -0·03 to 0·11; p=0·26]). There were no intervention-related serious adverse events. INTERPRETATION This kick and kill approach conferred no significant benefit compared with ART alone on measures of the HIV reservoir. Although this does not disprove the efficacy kick and kill strategy, for future trials enhancement of both kick and kill agents will be required. FUNDING Medical Research Council (MR/L00528X/1).
Collapse
Affiliation(s)
- Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, UK; NIHR Imperial Biomedical Research Centre, London, UK.
| | - Wolfgang Stöhr
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Matt Pace
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine Oxford Martin School, Oxford, UK
| | - Lucy Dorrell
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Andrew Lever
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sarah Pett
- Medical Research Council Clinical Trials Unit, University College London, London, UK; Institute for Global Health, University College London, London, UK; Mortimer Market Centre, London, UK
| | - Sabine Kinloch-de Loes
- Department of Infection and Immunity, Royal Free Hospital, London, UK; University College London, London, UK
| | - Julie Fox
- Department of Genitourinary Medicine and Infectious Disease, Guys and St Thomas' NHS Trust, London, UK; Department of Genitourinary Medicine and Infectious Disease, NIHR Biomedical Research Centre, Kings College London, London, UK
| | - Amanda Clarke
- Elton John Centre, Brighton, UK; Department of HIV and Sexual Health, Sussex University Hospital, Brighton, UK; Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Mark Nelson
- Chelsea and Westminster Hospital, Department of HIV Medicine, Imperial College London London, UK
| | - John Thornhill
- Department of Infectious Disease, Imperial College London, London, UK; NIHR Imperial Biomedical Research Centre, London, UK
| | - Maryam Khan
- Department of Infectious Disease, Imperial College London, London, UK; NIHR Imperial Biomedical Research Centre, London, UK
| | - Axel Fun
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Jakub Kopycinski
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Tomáš Hanke
- Nuffield Department of Medicine, Oxford University, UK; International Research Center for Medical Sciences, Kumamoto University, Japan
| | - Hongbing Yang
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Rachel Bennett
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | | | - Bonnie Howell
- Department of Infectious Disease and Vaccines, Merck and Co, West Point, PA, USA
| | - Richard Barnard
- Global Regulatory Affairs and Clinical Safety, Merck and Co, North Wales, PA, USA
| | - Guoxin Wu
- Department of Infectious Disease and Vaccines, Merck and Co, West Point, PA, USA
| | - Steve Kaye
- Department of Infectious Disease, Imperial College London, London, UK
| | - Mark Wills
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Abdel Babiker
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - John Frater
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
32
|
Steel R. Reconceptualising risk-benefit analyses: the case of HIV cure research. JOURNAL OF MEDICAL ETHICS 2020; 46:212-219. [PMID: 31732681 DOI: 10.1136/medethics-2019-105548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/25/2019] [Accepted: 10/08/2019] [Indexed: 06/10/2023]
Abstract
Modern antiretroviral therapies (ART) are capable of suppressing HIV in the bloodstream to undetectable levels. Nonetheless, people living with HIV must maintain lifelong adherence to ART to avoid the re-emergence of the infection. So despite the existence and efficacy of ART, there is still substantial interest in development of a cure. But HIV cure trials can be risky, their success is as of yet unlikely, and the medical gain of being cured is limited against a baseline of ART access. The medical prospect associated with participation in cure research thus look poor. Are the risks and burdens that HIV cure research places on participants so high that it is unethical, at present, to conduct it? In this paper, I answer 'no'. I start my argument by describing a foundational way of thinking about the ethical justification for regulatory limits on research risk; I then apply this way of thinking to HIV cure trials. In offering this analysis, I confine my attention to studies enrolling competent adults and I also do not consider risks research may pose to third parties or society. Rather, my concern is to engage with the thought that some trials are so risky that performing them is an ethically unacceptable way to treat the participants themselves. I reject this thought and instead argue that there is no level of risk, no matter how high, that inherently mistreats a participant.
Collapse
Affiliation(s)
- Robert Steel
- Clinical Center Department of Bioethics, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Abstract
The Berlin patient, a famous example for human immunodeficiency virus (HIV) cure, had received a bone marrow transplantation with an HIV resistance mutation. The authors describe his case and others that had shown HIV control, like the Mississippi baby who was started on antiretroviral therapy very early after birth, and posttreatment controllers, like the VISCONTI cohort. Moreover, the authors outline various strategies, oftentimes informed by these individuals, that have been tried in vitro, in animal models, or in human trials, to deplete the latent reservoir, which is considered the basis of HIV persistence and the obstacle to cure.
Collapse
Affiliation(s)
- Nikolaus Jilg
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Jonathan Z Li
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Bellizzi A, Ahye N, Jalagadugula G, Wollebo HS. A Broad Application of CRISPR Cas9 in Infectious Diseases of Central Nervous System. J Neuroimmune Pharmacol 2019; 14:578-594. [PMID: 31512166 PMCID: PMC6898781 DOI: 10.1007/s11481-019-09878-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/26/2019] [Indexed: 12/16/2022]
Abstract
Virus-induced diseases or neurological complications are huge socio-economic burden to human health globally. The complexity of viral-mediated CNS pathology is exacerbated by reemergence of new pathogenic neurotropic viruses of high public relevance. Although the central nervous system is considered as an immune privileged organ and is mainly protected by barrier system, there are a vast majority of neurotropic viruses capable of gaining access and cause diseases. Despite continued growth of the patient population and a number of treatment strategies, there is no successful viral specific therapy available for viral induced CNS diseases. Therefore, there is an urgent need for a clear alternative treatment strategy that can effectively target neurotropic viruses of DNA or RNA genome. To address this need, rapidly growing gene editing technology based on CRISPR/Cas9, provides unprecedented control over viral genome editing and will be an effective, highly specific and versatile tool for targeting CNS viral infection. In this review, we discuss the application of this system to control CNS viral infection and associated neurological disorders and future prospects. Graphical Abstract CRISPR/Cas9 technology as agent control over CNS viral infection.
Collapse
Affiliation(s)
- Anna Bellizzi
- Center for Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Nicholas Ahye
- Center for Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Gauthami Jalagadugula
- Center for Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hassen S Wollebo
- Center for Neurovirology, Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Room 756 MERB, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
35
|
Pinkevych M, Fennessey CM, Cromer D, Reid C, Trubey CM, Lifson JD, Keele BF, Davenport MP. Predictors of SIV recrudescence following antiretroviral treatment interruption. eLife 2019; 8:e49022. [PMID: 31650954 PMCID: PMC6917497 DOI: 10.7554/elife.49022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/24/2019] [Indexed: 12/26/2022] Open
Abstract
There is currently a need for proxy measures of the HIV rebound competent reservoir (RCR) that can predict viral rebound after combined antiretroviral treatment (cART) interruption. In this study, macaques infected with a barcoded SIVmac239 virus received cART beginning between 4- and 27 days post-infection, leading to the establishment of different levels of viral dissemination and persistence. Later treatment initiation led to higher SIV DNA levels maintained during treatment, which was significantly associated with an increased frequency of SIV reactivation and production of progeny capable of causing rebound viremia following treatment interruption. However, a 100-fold increase in SIV DNA in PBMCs was associated with only a 2-fold increase in the frequency of reactivation. These data suggest that the RCR can be established soon after infection, and that a large fraction of persistent viral DNA that accumulates after this time makes relatively little contribution to viral rebound.
Collapse
Affiliation(s)
- Mykola Pinkevych
- Infection Analytics ProgramKirby Institute for Infection and Immunity, UNSW AustraliaSydneyAustralia
| | - Christine M Fennessey
- AIDS and Cancer Virus ProgramFrederick National Laboratory for Cancer ResearchFrederickUnited States
| | - Deborah Cromer
- Infection Analytics ProgramKirby Institute for Infection and Immunity, UNSW AustraliaSydneyAustralia
| | - Carolyn Reid
- AIDS and Cancer Virus ProgramFrederick National Laboratory for Cancer ResearchFrederickUnited States
| | - Charles M Trubey
- AIDS and Cancer Virus ProgramFrederick National Laboratory for Cancer ResearchFrederickUnited States
| | - Jeffrey D Lifson
- AIDS and Cancer Virus ProgramFrederick National Laboratory for Cancer ResearchFrederickUnited States
| | - Brandon F Keele
- AIDS and Cancer Virus ProgramFrederick National Laboratory for Cancer ResearchFrederickUnited States
| | - Miles P Davenport
- Infection Analytics ProgramKirby Institute for Infection and Immunity, UNSW AustraliaSydneyAustralia
| |
Collapse
|
36
|
Kim GB, Hege K, Riley JL. CAR Talk: How Cancer-Specific CAR T Cells Can Instruct How to Build CAR T Cells to Cure HIV. Front Immunol 2019; 10:2310. [PMID: 31611880 PMCID: PMC6776630 DOI: 10.3389/fimmu.2019.02310] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/12/2019] [Indexed: 01/21/2023] Open
Abstract
Re-directing T cells via chimeric antigen receptors (CARs) was first tested in HIV-infected individuals with limited success, but these pioneering studies laid the groundwork for the clinically successful CD19 CARs that were recently FDA approved. Now there is great interest in revisiting the concept of using CAR-expressing T cells as part of a strategy to cure HIV. Many lessons have been learned on how to best engineer T cells to cure cancer, but not all of these lessons apply when developing CARs to treat and cure HIV. This mini review will focus on how early CAR T cell studies in HIV paved the way for cancer CAR T cell therapy and how progress in cancer CAR therapy has and will continue to be instructive for the development of HIV CAR T cell therapy. Additionally, the unique challenges that must be overcome to develop a successful HIV CAR T cell therapy will be highlighted.
Collapse
Affiliation(s)
- Gloria B. Kim
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kristen Hege
- Celgene Corporation, San Francisco, CA, United States
| | - James L. Riley
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
37
|
Huiting ED, Gittens K, Justement JS, Shi V, Blazkova J, Benko E, Kovacs C, Wender PA, Moir S, Sneller MC, Fauci AS, Chun TW. Impact of Treatment Interruption on HIV Reservoirs and Lymphocyte Subsets in Individuals Who Initiated Antiretroviral Therapy During the Early Phase of Infection. J Infect Dis 2019; 220:270-274. [PMID: 30840763 PMCID: PMC6941494 DOI: 10.1093/infdis/jiz100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/05/2019] [Indexed: 12/26/2022] Open
Abstract
Therapeutic strategies for achieving sustained virologic remission are being explored in human immunodeficiency virus (HIV)-infected individuals who began antiretroviral therapy (ART) during the early phase of infection. In the evaluation of such therapies, clinical protocols should include analytical treatment interruption (ATI); however, the immunologic and virologic impact of ATI in individuals who initiated ART early has not been fully delineated. We demonstrate that ATI causes neither expansion of HIV reservoirs nor immunologic abnormalities following reinitiation of ART. Our findings support the use of ATI to determine whether sustained virologic remission has been achieved in clinical trials of individuals who initiated ART early during HIV infection.
Collapse
Affiliation(s)
- Erin D Huiting
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | - Kathleen Gittens
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - J Shawn Justement
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | - Victoria Shi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | - Jana Blazkova
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | | | | | | | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | - Michael C Sneller
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | - Anthony S Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases
| |
Collapse
|
38
|
Panfil AR, London JA, Green PL, Yoder KE. CRISPR/Cas9 Genome Editing to Disable the Latent HIV-1 Provirus. Front Microbiol 2018; 9:3107. [PMID: 30619186 PMCID: PMC6302043 DOI: 10.3389/fmicb.2018.03107] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022] Open
Abstract
HIV-1 infection can be successfully controlled with anti-retroviral therapy (ART), but is not cured. A reservoir of cells harboring transcriptionally silent integrated provirus is able to reestablish replicating infection if ART is stopped. Latently HIV-1 infected cells are rare, but may persist for decades. Several novel strategies have been proposed to reduce the latent reservoir, including DNA sequence targeted CRISPR/Cas9 genome editing of the HIV-1 provirus. A significant challenge to genome editing is the sequence diversity of HIV-1 quasispecies present in patients. The high level of quasispecies diversity will require targeting of multiple sites in the viral genome and personalized engineering of a CRISPR/Cas9 regimen. The challenges of CRISPR/Cas9 delivery to the rare latently infected cells and quasispecies sequence diversity suggest that effective genome editing of every provirus is unlikely. However, recent evidence from post-treatment controllers, patients with controlled HIV-1 viral burden following interruption of ART, suggests a correlation between a reduced number of intact proviral sequences and control of the virus. The possibility of reducing the intact proviral sequences in patients by a genome editing technology remains intriguing, but requires significant advances in delivery to infected cells and identification of effective target sites.
Collapse
Affiliation(s)
- Amanda R. Panfil
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, United States
| | - James A. London
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Patrick L. Green
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, United States
| | - Kristine E. Yoder
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, United States
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
39
|
Veenhuis RT, Kwaa AK, Garliss CC, Latanich R, Salgado M, Pohlmeyer CW, Nobles CL, Gregg J, Scully EP, Bailey JR, Bushman FD, Blankson JN. Long-term remission despite clonal expansion of replication-competent HIV-1 isolates. JCI Insight 2018; 3:122795. [PMID: 30232278 DOI: 10.1172/jci.insight.122795] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/07/2018] [Indexed: 11/17/2022] Open
Abstract
Clonal expansion of T cells harboring replication-competent virus has recently been demonstrated in patients on suppressive antiretroviral therapy (ART) regimens. However, there has not been direct evidence of this phenomenon in settings of natural control, including in posttreatment controllers who maintain control of viral replication after treatment when ART is discontinued. We present a case of an individual who has had undetectable viral loads for more than 15 years following the cessation of ART. Using near-full-genome sequence analysis, we demonstrate that 9 of 12 replication-competent isolates cultured from this subject were identical and that this identity was maintained 6 months later. A similar pattern of replication-competent virus clonality was seen in a treatment-naive HLA-B*57 elite controller. In both cases, we show that CD8+ T cells are capable of suppressing the replication of the clonally expanded viruses in vitro. Our data suggest that, while clonal expansion of replication-competent virus can present a barrier to viral eradication, these viral isolates remain susceptible to HIV-specific immune responses and can be controlled in patients with long-term suppression of viral replication.
Collapse
Affiliation(s)
- Rebecca T Veenhuis
- Department of Medicine and.,Department of Molecular and Comparative Pathobiology, Center for AIDS Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | - Christopher L Nobles
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - John Gregg
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joel N Blankson
- Department of Medicine and.,Department of Molecular and Comparative Pathobiology, Center for AIDS Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|