1
|
Gay CL, Keys J, Kuruc JD, Sponaugle A, McGee KS, Ang C, Baker CE, Weimer ET, Schmitz JL, Archin NM, McKellar MS, Goonetilleke N, Margolis DM, Eron JJ. Dolutegravir/Abacavir/Lamivudine in Acute HIV-1 Results in Rapid Suppression and Restoration of CD4 T-cell Subsets Without Accelerated Decay of Latent HIV-1. Open Forum Infect Dis 2025; 12:ofaf247. [PMID: 40390705 PMCID: PMC12086330 DOI: 10.1093/ofid/ofaf247] [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: 10/18/2024] [Accepted: 04/22/2025] [Indexed: 05/21/2025] Open
Abstract
Background We evaluated rapid start of integrase-based antiretroviral therapy (ART) during acute HIV-1. Methods Adult participants initiated co-formulated dolutegravir/abacavir/lamivudine within 30 days of acute HIV-1 diagnosis. HLA-B*57-positive participants were excluded by rapid, flow cytometry screening. We evaluated HIV-1 RNA levels, CD4+ T-cell subsets, and change in replication competent HIV-1. Results Forty adults screened with 3 excluded due to positive HLA-B*57:01 or hepatitis B surface antigen results. All 37 participants starting study treatment suppressed to <200 copies/mL by week 24 (median of 4 weeks, interquartile range 3.4-5.1); 86% and 95% were <50 copies/mL at weeks 48 and 96, respectively. We observed a median 2.4-fold decline in frequency of resting CD4+ T-cell infection in a subset of participants providing 96 week samples. ART in acute HIV-1 resulted in CD4+ T-cell memory subpopulations similar to people without HIV-1 and preserved CD4+ and CD8+ T-cell frequencies compared to people starting ART in chronic HIV. Thirty-four participants required rapid HLA-B*57 testing at screening; 97% resulted ≤24 hours, and 71% started ART ≤24 hours. Conclusions Integrase-based ART during acute HIV-1 resulted in brisk viral suppression, preservation of CD4+ T-cell subsets, and decline in resting CD4+ T-cell infection.
Collapse
Affiliation(s)
- Cynthia L Gay
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- UNC Chapel Hill HIV Cure Center, University of North Carolina at Chapel Hilll, Chapel Hill, North Carolina, USA
| | - Jessica Keys
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
| | - JoAnn D Kuruc
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- UNC Chapel Hill HIV Cure Center, University of North Carolina at Chapel Hilll, Chapel Hill, North Carolina, USA
| | - Alexis Sponaugle
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kara S McGee
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Chelston Ang
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- UNC NC State Joint Department of Biomedical Engineering, Chapel Hill, North Carolina, USA
| | - Caroline E Baker
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
| | - Eric T Weimer
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John L Schmitz
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nancie M Archin
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- UNC Chapel Hill HIV Cure Center, University of North Carolina at Chapel Hilll, Chapel Hill, North Carolina, USA
| | - Mehri S McKellar
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Nilu Goonetilleke
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- UNC Chapel Hill HIV Cure Center, University of North Carolina at Chapel Hilll, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David M Margolis
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- UNC Chapel Hill HIV Cure Center, University of North Carolina at Chapel Hilll, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill (UNC) School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Reeves DB, Litchford M, Fish CS, Farrell-Sherman A, Poindexter M, Ahmed N, Cassidy NAJ, Neary J, Wamalwa D, Langat A, Chebet D, Moraa H, Antar AAR, Slyker J, Benki-Nugent S, Cohn LB, Schiffer JT, Overbaugh J, John-Stewart G, Lehman DA. Intact HIV DNA decays in children with and without complete viral load suppression. PLoS Pathog 2025; 21:e1013003. [PMID: 40184428 PMCID: PMC12002518 DOI: 10.1371/journal.ppat.1013003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/16/2025] [Accepted: 02/21/2025] [Indexed: 04/06/2025] Open
Abstract
To inform cure in children living with HIV (CWH), we elucidated the dynamics and mechanisms underlying HIV persistence during antiretroviral therapy (ART). In 120 Kenyan CWH who initiated ART between 1-12 months of age, 55 had durable viral load suppression, and 65 experienced ART interruptions. We measured plasma HIV RNA levels, CD4+ T cell count, and levels of intact and defective HIV DNA proviruses via the cross-subtype intact proviral DNA assay (CS-IPDA). By modeling data from the durably suppressed subset, we found that during early ART (year 0-1 on ART), plasma RNA levels decayed rapidly and biphasically and intact and defective HIV DNA decayed with mean 3 and 9 month half-lives, respectively. After viral suppression was achieved (years 1-8 on ART), intact HIV DNA decay slowed to a mean 22 month half-life, whilst defective HIV DNA no longer decayed. In five CWH, we found individual CD4+ TCRβ clones wax and wane, but average kinetics resembled those of defective DNA and CD4 count, suggesting that differential decay of intact HIV DNA arises from selective pressures overlaying normal CD4+ T cell kinetics. Finally, by modeling HIV RNA and DNA in CWH with treatment interruptions, we linked temporary viremia to transient rises in HIV DNA, but long-term intact reservoirs were not strongly influenced, suggesting brief treatment interruptions may not significantly increase HIV reservoirs in children.
Collapse
Affiliation(s)
- Daniel B Reeves
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Global Health, University of Washington, Seattle, Washington, United States of America
| | - Morgan Litchford
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Carolyn S Fish
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Anna Farrell-Sherman
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Makayla Poindexter
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Nashwa Ahmed
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Noah A J Cassidy
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Jillian Neary
- Global Health, University of Washington, Seattle, Washington, United States of America
| | - Dalton Wamalwa
- Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Agnes Langat
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Daisy Chebet
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Hellen Moraa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Annukka A R Antar
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jennifer Slyker
- Global Health, University of Washington, Seattle, Washington, United States of America
| | - Sarah Benki-Nugent
- Global Health, University of Washington, Seattle, Washington, United States of America
| | - Lillian B Cohn
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Joshua T Schiffer
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Julie Overbaugh
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Grace John-Stewart
- Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Dara A Lehman
- Global Health, University of Washington, Seattle, Washington, United States of America
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
3
|
Farinre O, Anaya T, King AC, Endrias K, Hébert AH, Hill AL, Jean S, Wood JS, Ehnert S, Liang S, Laird GM, Mason RD, Roederer M, Safrit JT, Mavigner M, Chahroudi A. SIV Env RhmAbs + N-803 at ART initiation prolongs viral decay without disrupting reservoir establishment in SIV-infected infant macaques. PLoS Pathog 2025; 21:e1012863. [PMID: 39792949 PMCID: PMC11756789 DOI: 10.1371/journal.ppat.1012863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/23/2025] [Accepted: 12/26/2024] [Indexed: 01/12/2025] Open
Abstract
The latent viral reservoir remains the major barrier to HIV cure, placing the burden of strict adherence to antiretroviral therapy (ART) on people living with HIV to prevent recrudescence of viremia. For infants with perinatally acquired HIV, adherence is anticipated to be a lifelong need. In this study, we tested the hypothesis that administration of ART and viral Envelope-specific rhesus-derived IgG1 monoclonal antibodies (RhmAbs) with or without the IL-15 superagonist N-803 early in infection would limit viral reservoir establishment in SIV-infected infant rhesus macaques. Following initiation of ART at 1-2 weeks after oral SIVmac251 infection, we observed biphasic decay of viremia, with first phase decay significantly faster in the ART + SIV RhmAbs-treated group compared to controls that received only ART. In contrast, the addition of N-803 to ART + SIV RhmAbs significantly slowed both the first and second phase viral decay compared to the ART only group. Treatment with a single dose of N-803 resulted in increased frequency of Ki67 expressing NK, CD8+, and CD4+ T cells. Levels of intact SIV proviruses in CD4+ T cells from blood, lymph nodes, and rectum at week 48 of ART did not differ across groups. Similarly, the time to viral rebound following ART interruption was not impacted by the experimental treatments. These results support the concept that the rebound-competent viral reservoir is formed within days after infection and that targeting only productively infected cells for clearance near the time of ART initiation, even during acute infection, may be insufficient to limit reservoir establishment.
Collapse
Affiliation(s)
- Omotayo Farinre
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Tzoalli Anaya
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Alexis C. King
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kedan Endrias
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Anne H. Hébert
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alison L. Hill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sherrie Jean
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jennifer S. Wood
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Shan Liang
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Gregory M. Laird
- Accelevir Diagnostics, Baltimore, Maryland, United States of America
| | - Rosemarie D. Mason
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Mario Roederer
- ImmunoTechnology Section, National Institutes of Allergy and Infectious Diseases, Bethesda, Massachusetts, United States of America
| | | | - Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, United States of America
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
4
|
González-Navarro I, Urrea V, Gálvez C, Garcia-Guerrero MDC, Morón-López S, Puertas MC, Grau E, Mothe B, Bailón L, Miranda C, García F, Leal L, Vandekerckhove L, Marconi VC, Sekaly RP, Clotet B, Martinez-Picado J, Salgado M. Assessing advances in three decades of clinical antiretroviral therapy on the HIV-1 reservoir. J Clin Invest 2024; 135:e183952. [PMID: 39610346 PMCID: PMC11735095 DOI: 10.1172/jci183952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUNDAntiretroviral therapy (ART) has improved the clinical management of HIV-1 infection. However, little is known about how the latest ART recommendations affect the heterogeneity of the HIV-1 reservoir size.METHODSWe used a complete statistical approach to outline parameters underlying the diversity in HIV-1 reservoir size in a cohort of 892 people with HIV-1 (PWH) on suppressive ART for more than 3 years. Total HIV-1-DNA levels were measured in PBMCs using digital droplet PCR (ddPCR).RESULTSWe classified 179 (20%) participants as being low viral reservoir treated (LoViReT) (<50 HIV-1-DNA copies/106 PBMCs). Twenty variables were collected to explore their association with the LoViReT phenotype using machine learning approaches. LoViReT status was closely associated with higher nadir CD4, lower zenith pre-ART viral load, lower CD4 recovery, shorter time from diagnosis to undetectable viral load, and initiation of treatment with an integrase inhibitor-containing (InSTI-containing) regimen. Initiation of ART with any InSTI was also linked with a shorter time to undetectable viremia. Locally estimated scatterplot smoothing (LOESS) regression revealed a progressive reduction in the size of the HIV-1 reservoir in individuals who started ART after 2007. Similarly, a higher nadir CD4 and a shorter time to undetectable viremia were observed when treatment was initiated after that year.CONCLUSIONOur findings demonstrate that the progressive implementation of earlier, universal treatment at diagnosis and the use of InSTIs affected the size of the HIV-1 reservoir. Our work shows that effective management of infection is the first step toward reducing the reservoir and brings us closer to achieving a cure.FUNDINGNIH; Division of AIDS at the National Institute of Allergy and Infectious Diseases (NIAID), NIH; Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Irene González-Navarro
- IrsiCaixa, Badalona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | | | | | | | - Sara Morón-López
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
| | - Maria C. Puertas
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
| | | | - Beatriz Mothe
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
- Chair in Infectious Diseases and Immunity, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Lucía Bailón
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Cristina Miranda
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
| | - Felipe García
- Infectious Diseases Department Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Lorna Leal
- Infectious Diseases Department Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Vincent C. Marconi
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
- The Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Rafick P. Sekaly
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
- Pathology Advanced Translational Research Unit (PATRU), Department of Pathology and Laboratory Medicine and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bonaventura Clotet
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
- Chair in Infectious Diseases and Immunity, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Javier Martinez-Picado
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
- Chair in Infectious Diseases and Immunity, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Maria Salgado
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
| | | |
Collapse
|
5
|
Sponaugle A, Weideman AMK, Ranek J, Atassi G, Kuruc J, Adimora AA, Archin NM, Gay C, Kuritzkes DR, Margolis DM, Vincent BG, Stanley N, Hudgens MG, Eron JJ, Goonetilleke N. Dominant CD4 + T cell receptors remain stable throughout antiretroviral therapy-mediated immune restoration in people with HIV. Cell Rep Med 2023; 4:101268. [PMID: 37949070 PMCID: PMC10694675 DOI: 10.1016/j.xcrm.2023.101268] [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: 01/12/2023] [Revised: 06/05/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
In people with HIV (PWH), the post-antiretroviral therapy (ART) window is critical for immune restoration and HIV reservoir stabilization. We employ deep immune profiling and T cell receptor (TCR) sequencing and examine proliferation to assess how ART impacts T cell homeostasis. In PWH on long-term ART, lymphocyte frequencies and phenotypes are mostly stable. By contrast, broad phenotypic changes in natural killer (NK) cells, γδ T cells, B cells, and CD4+ and CD8+ T cells are observed in the post-ART window. Whereas CD8+ T cells mostly restore, memory CD4+ T subsets and cytolytic NK cells show incomplete restoration 1.4 years post ART. Surprisingly, the hierarchies and frequencies of dominant CD4 TCR clonotypes (0.1%-11% of all CD4+ T cells) remain stable post ART, suggesting that clonal homeostasis can be independent of homeostatic processes regulating CD4+ T cell absolute number, phenotypes, and function. The slow restoration of host immunity post ART also has implications for the design of ART interruption studies.
Collapse
Affiliation(s)
- Alexis Sponaugle
- Department of Microbiology & Immunology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Ann Marie K Weideman
- Department of Biostatistics, UNC Chapel Hill, Chapel Hill, NC, USA; Center for AIDS Research, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Jolene Ranek
- Computational Medicine Program, UNC Chapel Hill, Chapel Hill, NC, USA; Curriculum in Bioinformatics and Computational Biology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Gatphan Atassi
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - JoAnn Kuruc
- Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Adaora A Adimora
- Center for AIDS Research, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA; Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Nancie M Archin
- Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Cynthia Gay
- Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David M Margolis
- Department of Microbiology & Immunology, UNC Chapel Hill, Chapel Hill, NC, USA; Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Benjamin G Vincent
- Department of Microbiology & Immunology, UNC Chapel Hill, Chapel Hill, NC, USA; Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA; Curriculum in Bioinformatics and Computational Biology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Natalie Stanley
- Computational Medicine Program, UNC Chapel Hill, Chapel Hill, NC, USA; Department of Computer Science, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Michael G Hudgens
- Department of Biostatistics, UNC Chapel Hill, Chapel Hill, NC, USA; Center for AIDS Research, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Joseph J Eron
- Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Nilu Goonetilleke
- Department of Microbiology & Immunology, UNC Chapel Hill, Chapel Hill, NC, USA; Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
6
|
Policicchio BB, Cardozo-Ojeda EF, Xu C, Ma D, He T, Raehtz KD, Sivanandham R, Kleinman AJ, Perelson AS, Apetrei C, Pandrea I, Ribeiro RM. CD8 + T cells control SIV infection using both cytolytic effects and non-cytolytic suppression of virus production. Nat Commun 2023; 14:6657. [PMID: 37863982 PMCID: PMC10589330 DOI: 10.1038/s41467-023-42435-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/11/2023] [Indexed: 10/22/2023] Open
Abstract
Whether CD8+ T lymphocytes control human immunodeficiency virus infection by cytopathic or non-cytopathic mechanisms is not fully understood. Multiple studies highlighted non-cytopathic effects, but one hypothesis is that cytopathic effects of CD8+ T cells occur before viral production. Here, to examine the role of CD8+ T cells prior to virus production, we treated SIVmac251-infected macaques with an integrase inhibitor combined with a CD8-depleting antibody, or with either reagent alone. We analyzed the ensuing viral dynamics using a mathematical model that included infected cells pre- and post- viral DNA integration to compare different immune effector mechanisms. Macaques receiving the integrase inhibitor alone experienced greater viral load decays, reaching lower nadirs on treatment, than those treated also with the CD8-depleting antibody. Models including CD8+ cell-mediated reduction of viral production (non-cytolytic) were found to best explain the viral profiles across all macaques, in addition an effect in killing infected cells pre-integration (cytolytic) was supported in some of the best models. Our results suggest that CD8+ T cells have both a cytolytic effect on infected cells before viral integration, and a direct, non-cytolytic effect by suppressing viral production.
Collapse
Affiliation(s)
- Benjamin B Policicchio
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Dongzhu Ma
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Tianyu He
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Kevin D Raehtz
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ranjit Sivanandham
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Adam J Kleinman
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ivona Pandrea
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA.
- Laboratório de Biomatemática, Faculdade de Medicina da Universidade de Lisboa (previous address), Lisboa, Portugal.
| |
Collapse
|
7
|
Cody JW, Ellis-Connell AL, O’Connor SL, Pienaar E. Mathematical modeling indicates that regulatory inhibition of CD8+ T cell cytotoxicity can limit efficacy of IL-15 immunotherapy in cases of high pre-treatment SIV viral load. PLoS Comput Biol 2023; 19:e1011425. [PMID: 37616311 PMCID: PMC10482305 DOI: 10.1371/journal.pcbi.1011425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/06/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Immunotherapeutic cytokines can activate immune cells against cancers and chronic infections. N-803 is an IL-15 superagonist that expands CD8+ T cells and increases their cytotoxicity. N-803 also temporarily reduced viral load in a limited subset of non-human primates infected with simian immunodeficiency virus (SIV), a model of HIV. However, viral suppression has not been observed in all SIV cohorts and may depend on pre-treatment viral load and the corresponding effects on CD8+ T cells. Starting from an existing mechanistic mathematical model of N-803 immunotherapy of SIV, we develop a model that includes activation of SIV-specific and non-SIV-specific CD8+ T cells by antigen, inflammation, and N-803. Also included is a regulatory counter-response that inhibits CD8+ T cell proliferation and function, representing the effects of immune checkpoint molecules and immunosuppressive cells. We simultaneously calibrate the model to two separate SIV cohorts. The first cohort had low viral loads prior to treatment (≈3-4 log viral RNA copy equivalents (CEQ)/mL), and N-803 treatment transiently suppressed viral load. The second had higher pre-treatment viral loads (≈5-7 log CEQ/mL) and saw no consistent virus suppression with N-803. The mathematical model can replicate the viral and CD8+ T cell dynamics of both cohorts based on different pre-treatment viral loads and different levels of regulatory inhibition of CD8+ T cells due to those viral loads (i.e. initial conditions of model). Our predictions are validated by additional data from these and other SIV cohorts. While both cohorts had high numbers of activated SIV-specific CD8+ T cells in simulations, viral suppression was precluded in the high viral load cohort due to elevated inhibition of cytotoxicity. Thus, we mathematically demonstrate how the pre-treatment viral load can influence immunotherapeutic efficacy, highlighting the in vivo conditions and combination therapies that could maximize efficacy and improve treatment outcomes.
Collapse
Affiliation(s)
- Jonathan W. Cody
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
8
|
Roux HM, Figueiredo S, Sareoua L, Salmona M, Hamroune J, Adoux L, Migraine J, Hance A, Clavel F, Cheynier R, Dutrieux J. DNA ultra-sensitive quantification, a technology for studying HIV unintegrated linear DNA. CELL REPORTS METHODS 2023; 3:100443. [PMID: 37159665 PMCID: PMC10162948 DOI: 10.1016/j.crmeth.2023.100443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 01/28/2023] [Accepted: 03/10/2023] [Indexed: 05/11/2023]
Abstract
Unintegrated HIV DNA represents between 20% and 35% of the total viral DNA in infected patients. Only the linear forms (unintegrated linear DNAs [ULDs]) can be substrates for integration and for the completion of a full viral cycle. In quiescent cells, these ULDs may be responsible for pre-integrative latency. However, their detection remains difficult due to the lack of specificity and sensitivity of existing techniques. We developed an ultra-sensitive, specific, and high-throughput technology for ULD quantification called DUSQ (DNA ultra-sensitive quantification) combining linker-mediated PCR and next-generation sequencing (NGS) using molecular barcodes. Studying cells with different activity levels, we determined that the ULD half-life goes up to 11 days in resting CD4+ T cells. Finally, we were able to quantify ULDs in samples from patients infected with HIV-1, providing a proof of concept for the use of DUSQ in vivo to track pre-integrative latency. DUSQ can be adapted to the detection of other rare DNA molecules.
Collapse
Affiliation(s)
- Hélène Marie Roux
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
| | - Suzanne Figueiredo
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
| | - Lucas Sareoua
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
| | - Maud Salmona
- Université Paris Cité, Paris, France
- INSERM U976, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Saint Louis, Laboratoire de Virologie, Paris, France
| | - Juliette Hamroune
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
| | - Lucie Adoux
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
| | | | | | - François Clavel
- Université Paris Cité, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Saint Louis, Laboratoire de Virologie, Paris, France
| | - Rémi Cheynier
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
| | - Jacques Dutrieux
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS, UMR8104, 75014 Paris, France
- Viral DNA Integration and Chromatin Dynamics Network (DyNAVir), France
- Corresponding author
| |
Collapse
|
9
|
Sass J, Awasthi A, Obregon-Perko V, McCarthy J, Lloyd AL, Chahroudi A, Permar S, Chan C. A simple model for viral decay dynamics and the distribution of infected cell life spans in SHIV-infected infant rhesus macaques. Math Biosci 2023; 356:108958. [PMID: 36567003 PMCID: PMC9918703 DOI: 10.1016/j.mbs.2022.108958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
The dynamics of HIV viral load following the initiation of antiretroviral therapy is not well-described by simple, single-phase exponential decay. Several mathematical models have been proposed to describe its more complex behavior, the most popular of which is two-phase exponential decay. The underlying assumption in two-phase exponential decay is that there are two classes of infected cells with different lifespans. However, with the exception of CD4+ T cells, there is not a consensus on all of the cell types that can become productively infected, and the fit of the two-phase exponential decay to observed data from SHIV.C.CH505 infected infant rhesus macaques was relatively poor. Therefore, we propose a new model for viral decay, inspired by the Gompertz model where the decay rate itself is a dynamic variable. We modify the Gompertz model to include a linear term that modulates the decay rate. We show that this simple model performs as well as the two-phase exponential decay model on HIV and SIV data sets, and outperforms it for the infant rhesus macaque SHIV.C.CH505 infection data set. We also show that by using a stochastic differential equation formulation, the modified Gompertz model can be interpreted as being driven by a population of infected cells with a continuous distribution of cell lifespans, and estimate this distribution for the SHIV.C.CH505-infected infant rhesus macaques. Thus, we find that the dynamics of viral decay in this model of infant HIV infection and treatment may be explained by a distribution of cell lifespans, rather than two distinct cell types.
Collapse
Affiliation(s)
- Julian Sass
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA.
| | - Achal Awasthi
- Department of Bioinformatics and Biostatistics, Duke University, Durham, USA; Duke Center for Human Systems Immunology, Duke University, Durham, USA.
| | | | - Janice McCarthy
- Department of Bioinformatics and Biostatistics, Duke University, Durham, USA; Duke Center for Human Systems Immunology, Duke University, Durham, USA.
| | - Alun L Lloyd
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA.
| | - Ann Chahroudi
- Department of Pediatrics, Emory University, Atlanta, USA; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, NY, USA
| | - Cliburn Chan
- Department of Bioinformatics and Biostatistics, Duke University, Durham, USA; Duke Center for Human Systems Immunology, Duke University, Durham, USA.
| |
Collapse
|
10
|
White JA, Simonetti FR, Beg S, McMyn NF, Dai W, Bachmann N, Lai J, Ford WC, Bunch C, Jones JL, Ribeiro RM, Perelson AS, Siliciano JD, Siliciano RF. Complex decay dynamics of HIV virions, intact and defective proviruses, and 2LTR circles following initiation of antiretroviral therapy. Proc Natl Acad Sci U S A 2022; 119:e2120326119. [PMID: 35110411 PMCID: PMC8833145 DOI: 10.1073/pnas.2120326119] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
In persons living with HIV-1 (PLWH) who start antiretroviral therapy (ART), plasma virus decays in a biphasic fashion to below the detection limit. The first phase reflects the short half-life (<1 d) of cells that produce most of the plasma virus. The second phase represents the slower turnover (t1/2 = 14 d) of another infected cell population, whose identity is unclear. Using the intact proviral DNA assay (IPDA) to distinguish intact and defective proviruses, we analyzed viral decay in 17 PLWH initiating ART. Circulating CD4+ T cells with intact proviruses include few of the rapidly decaying first-phase cells. Instead, this population initially decays more slowly (t1/2 = 12.9 d) in a process that largely represents death or exit from the circulation rather than transition to latency. This more protracted decay potentially allows for immune selection. After ∼3 mo, the decay slope changes, and CD4+ T cells with intact proviruses decay with a half-life of 19 mo, which is still shorter than that of the latently infected cells that persist on long-term ART. Two-long-terminal repeat (2LTR) circles decay with fast and slow phases paralleling intact proviruses, a finding that precludes their use as a simple marker of ongoing viral replication. Proviruses with defects at the 5' or 3' end of the genome show equivalent monophasic decay at rates that vary among individuals. Understanding these complex early decay processes is important for correct use of reservoir assays and may provide insights into properties of surviving cells that can constitute the stable latent reservoir.
Collapse
Affiliation(s)
- Jennifer A White
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Francesco R Simonetti
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Subul Beg
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Natalie F McMyn
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Weiwei Dai
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Niklas Bachmann
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jun Lai
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - William C Ford
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Christina Bunch
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Joyce L Jones
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ruy M Ribeiro
- Department of Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Alan S Perelson
- Department of Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Janet D Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205;
- HHMI, Baltimore, MD 21205
| |
Collapse
|
11
|
Goyal A, Gardner M, Mayer BT, Jerome KR, Farzan M, Schiffer JT, Cardozo-Ojeda EF. Estimation of the in vivo neutralization potency of eCD4Ig and conditions for AAV-mediated production for SHIV long-term remission. SCIENCE ADVANCES 2022; 8:eabj5666. [PMID: 35020436 PMCID: PMC8754410 DOI: 10.1126/sciadv.abj5666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/18/2021] [Indexed: 06/14/2023]
Abstract
The engineered protein eCD4Ig has emerged as a promising approach to achieve HIV remission in the absence of antiviral therapy. eCD4Ig neutralizes nearly all HIV-1 isolates and induces antibody-dependent cell-mediated cytotoxicity (ADCC) in vitro. To characterize the in vivo antiviral neutralization and possible ADCC effects of eCD4Ig, we fit mathematical models to eCD4Ig, anti–eCD4Ig-drug antibody (ADA), and viral load kinetics from healthy and simian-human immunodeficiency virus AD8 (SHIV-AD8) infected nonhuman primates that were treated with single or sequentially dosed eCD4Ig passive administrations. Our model predicts that eCD4Ig transiently decreases SHIV viral loads due to neutralization only with an in vivo IC50 of ~25 μg/ml but with limited effect due to ADA. Simulations suggest that endogenous, continuous expression of eCD4Ig at levels greater than 105 μg/day, as is possible with Adeno-associated virus (AAV) vector-based production, could overcome the diminishing effects of ADA and allow for long-term remission of SHIV viremia in nonhuman primates.
Collapse
Affiliation(s)
- Ashish Goyal
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Bryan T. Mayer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Keith R. Jerome
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael Farzan
- Department of Immunology and Microbiology, Scripps Research Institute, Florida Campus, Jupiter, FL, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - E. Fabian Cardozo-Ojeda
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
12
|
Lau CY, Adan MA, Maldarelli F. Why the HIV Reservoir Never Runs Dry: Clonal Expansion and the Characteristics of HIV-Infected Cells Challenge Strategies to Cure and Control HIV Infection. Viruses 2021; 13:2512. [PMID: 34960781 PMCID: PMC8708047 DOI: 10.3390/v13122512] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/22/2021] [Accepted: 11/27/2021] [Indexed: 12/13/2022] Open
Abstract
Antiretroviral therapy (ART) effectively reduces cycles of viral replication but does not target proviral populations in cells that persist for prolonged periods and that can undergo clonal expansion. Consequently, chronic human immunodeficiency virus (HIV) infection is sustained during ART by a reservoir of long-lived latently infected cells and their progeny. This proviral landscape undergoes change over time on ART. One of the forces driving change in the landscape is the clonal expansion of infected CD4 T cells, which presents a key obstacle to HIV eradication. Potential mechanisms of clonal expansion include general immune activation, antigenic stimulation, homeostatic proliferation, and provirus-driven clonal expansion, each of which likely contributes in varying, and largely unmeasured, amounts to maintaining the reservoir. The role of clinical events, such as infections or neoplasms, in driving these mechanisms remains uncertain, but characterizing these forces may shed light on approaches to effectively eradicate HIV. A limited number of individuals have been cured of HIV infection in the setting of bone marrow transplant; information from these and other studies may identify the means to eradicate or control the virus without ART. In this review, we describe the mechanisms of HIV-1 persistence and clonal expansion, along with the attempts to modify these factors as part of reservoir reduction and cure strategies.
Collapse
Affiliation(s)
- Chuen-Yen Lau
- HIV Dynamics and Replication Program, NCI, NIH, Bethesda, MD 20892, USA; (C.-Y.L.); (M.A.A.)
| | - Matthew A. Adan
- HIV Dynamics and Replication Program, NCI, NIH, Bethesda, MD 20892, USA; (C.-Y.L.); (M.A.A.)
- Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, NCI, NIH, Bethesda, MD 20892, USA; (C.-Y.L.); (M.A.A.)
| |
Collapse
|
13
|
Cardozo-Ojeda EF, Perelson AS. Modeling HIV-1 Within-Host Dynamics After Passive Infusion of the Broadly Neutralizing Antibody VRC01. Front Immunol 2021; 12:710012. [PMID: 34531859 PMCID: PMC8438300 DOI: 10.3389/fimmu.2021.710012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/02/2021] [Indexed: 11/20/2022] Open
Abstract
VRC01 is a broadly neutralizing antibody that targets the CD4 binding site of HIV-1 gp120. Passive administration of VRC01 in humans has assessed the safety and the effect on plasma viremia of this monoclonal antibody (mAb) in a phase 1 clinical trial. After VRC01 infusion, the plasma viral load in most of the participants was reduced but had particular dynamics not observed during antiretroviral therapy. In this paper, we introduce different mathematical models to explain the observed dynamics and fit them to the plasma viral load data. Based on the fitting results we argue that a model containing reversible Ab binding to virions and clearance of virus-VRC01 complexes by a two-step process that includes (1) saturable capture followed by (2) internalization/degradation by phagocytes, best explains the data. This model predicts that VRC01 may enhance the clearance of Ab-virus complexes, explaining the initial viral decay observed immediately after antibody infusion in some participants. Because Ab-virus complexes are assumed to be unable to infect cells, i.e., contain neutralized virus, the model predicts a longer-term viral decay consistent with that observed in the VRC01 treated participants. By assuming a homogeneous viral population sensitive to VRC01, the model provides good fits to all of the participant data. However, the fits are improved by assuming that there were two populations of virus, one more susceptible to antibody-mediated neutralization than the other.
Collapse
Affiliation(s)
- E Fabian Cardozo-Ojeda
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Alan S Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, United States
| |
Collapse
|
14
|
Cody JW, Ellis-Connell AL, O’Connor SL, Pienaar E. Mathematical modeling of N-803 treatment in SIV-infected non-human primates. PLoS Comput Biol 2021; 17:e1009204. [PMID: 34319980 PMCID: PMC8351941 DOI: 10.1371/journal.pcbi.1009204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 08/09/2021] [Accepted: 06/21/2021] [Indexed: 12/01/2022] Open
Abstract
Immunomodulatory drugs could contribute to a functional cure for Human Immunodeficiency Virus (HIV). Interleukin-15 (IL-15) promotes expansion and activation of CD8+ T cell and natural killer (NK) cell populations. In one study, an IL-15 superagonist, N-803, suppressed Simian Immunodeficiency Virus (SIV) in non-human primates (NHPs) who had received prior SIV vaccination. However, viral suppression attenuated with continued N-803 treatment, partially returning after long treatment interruption. While there is evidence of concurrent drug tolerance, immune regulation, and viral escape, the relative contributions of these mechanisms to the observed viral dynamics have not been quantified. Here, we utilize mathematical models of N-803 treatment in SIV-infected macaques to estimate contributions of these three key mechanisms to treatment outcomes: 1) drug tolerance, 2) immune regulation, and 3) viral escape. We calibrated our model to viral and lymphocyte responses from the above-mentioned NHP study. Our models track CD8+ T cell and NK cell populations with N-803-dependent proliferation and activation, as well as viral dynamics in response to these immune cell populations. We compared mathematical models with different combinations of the three key mechanisms based on Akaike Information Criterion and important qualitative features of the NHP data. Two minimal models were capable of reproducing the observed SIV response to N-803. In both models, immune regulation strongly reduced cytotoxic cell activation to enable viral rebound. Either long-term drug tolerance or viral escape (or some combination thereof) could account for changes to viral dynamics across long breaks in N-803 treatment. Theoretical explorations with the models showed that less-frequent N-803 dosing and concurrent immune regulation blockade (e.g. PD-L1 inhibition) may improve N-803 efficacy. However, N-803 may need to be combined with other immune therapies to countermand viral escape from the CD8+ T cell response. Our mechanistic model will inform such therapy design and guide future studies. Immune therapy may be a critical component in the functional cure for Human Immunodeficiency Virus (HIV). N-803 is an immunotherapeutic drug that activates antigen-specific CD8+ T cells of the immune system. These CD8+ T cells eliminate HIV-infected cells in order to limit the spread of infection in the body. In one study, N-803 reduced plasma viremia in macaques that were infected with Simian Immunodeficiency Virus, an analog of HIV. Here, we used mathematical models to analyze the data from this study to better understand the effects of N-803 therapy on the immune system. Our models indicated that inhibitory signals may be reversing the stimulatory effect of N-803. Results also suggested the possibilities that tolerance to N-803 could build up within the CD8+ T cells themselves and that the treatment may be selecting for virus strains that are not targeted by CD8+ T cells. Our models predict that N-803 therapy may be made more effective if the time between doses is increased or if inhibitory signals are blocked by an additional drug. Also, N-803 may need to be combined with other immune therapies to target virus that would otherwise evade CD8+ T cells.
Collapse
Affiliation(s)
- Jonathan W. Cody
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
15
|
Mechanistic basis of post-treatment control of SIV after anti-α4β7 antibody therapy. PLoS Comput Biol 2021; 17:e1009031. [PMID: 34106916 PMCID: PMC8189501 DOI: 10.1371/journal.pcbi.1009031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Treating macaques with an anti-α4β7 antibody under the umbrella of combination antiretroviral therapy (cART) during early SIV infection can lead to viral remission, with viral loads maintained at < 50 SIV RNA copies/ml after removal of all treatment in a subset of animals. Depletion of CD8+ lymphocytes in controllers resulted in transient recrudescence of viremia, suggesting that the combination of cART and anti-α4β7 antibody treatment led to a state where ongoing immune responses kept the virus undetectable in the absence of treatment. A previous mathematical model of HIV infection and cART incorporates immune effector cell responses and exhibits the property of two different viral load set-points. While the lower set-point could correspond to the attainment of long-term viral remission, attaining the higher set-point may be the result of viral rebound. Here we expand that model to include possible mechanisms of action of an anti-α4β7 antibody operating in these treated animals. We show that the model can fit the longitudinal viral load data from both IgG control and anti-α4β7 antibody treated macaques, suggesting explanations for the viral control associated with cART and an anti-α4β7 antibody treatment. This effective perturbation to the virus-host interaction can also explain observations in other nonhuman primate experiments in which cART and immunotherapy have led to post-treatment control or resetting of the viral load set-point. Interestingly, because the viral kinetics in the various treated animals differed—some animals exhibited large fluctuations in viral load after cART cessation—the model suggests that anti-α4β7 treatment could act by different primary mechanisms in different animals and still lead to post-treatment viral control. This outcome is nonetheless in accordance with a model with two stable viral load set-points, in which therapy can perturb the system from one set-point to a lower one through different biological mechanisms. Some macaques treated with an anti-α4β7 monoclonal antibody along with antiretroviral therapy during the early stages of simian immunodeficiency virus infection had their viral load become undetectable (below 50 SIV RNA copies/ml) after all treatment was stopped, whereas animals not given the antibody all had their viral loads rebound to high levels. Using a mathematical model, we examined four potential ways in which the antibody could have altered the balance between viral growth and immune control to maintain an undetectable viral load. We show that a shift to controlled infection can occur through multiple biologically reasonable mechanisms of action of the anti-α4β7 antibody.
Collapse
|
16
|
Cardozo-Ojeda EF, Duke ER, Peterson CW, Reeves DB, Mayer BT, Kiem HP, Schiffer JT. Thresholds for post-rebound SHIV control after CCR5 gene-edited autologous hematopoietic cell transplantation. eLife 2021; 10:e57646. [PMID: 33432929 PMCID: PMC7803377 DOI: 10.7554/elife.57646] [Citation(s) in RCA: 12] [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: 04/07/2020] [Accepted: 12/27/2020] [Indexed: 01/10/2023] Open
Abstract
Autologous, CCR5 gene-edited hematopoietic stem and progenitor cell (HSPC) transplantation is a promising strategy for achieving HIV remission. However, only a fraction of HSPCs can be edited ex vivo to provide protection against infection. To project the thresholds of CCR5-edition necessary for HIV remission, we developed a mathematical model that recapitulates blood T cell reconstitution and plasma simian-HIV (SHIV) dynamics from SHIV-1157ipd3N4-infected pig-tailed macaques that underwent autologous transplantation with CCR5 gene editing. The model predicts that viral control can be obtained following analytical treatment interruption (ATI) when: (1) transplanted HSPCs are at least fivefold higher than residual endogenous HSPCs after total body irradiation and (2) the fraction of protected HSPCs in the transplant achieves a threshold (76-94%) sufficient to overcome transplantation-dependent loss of SHIV immunity. Under these conditions, if ATI is withheld until transplanted gene-modified cells engraft and reconstitute to a steady state, spontaneous viral control is projected to occur.
Collapse
Affiliation(s)
| | - Elizabeth R Duke
- Vaccine and Infectious Disease Division, University of WashingtonSeattleUnited States
- Department of Medicine, University of WashingtonSeattleUnited States
| | - Christopher W Peterson
- Department of Medicine, University of WashingtonSeattleUnited States
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Daniel B Reeves
- Vaccine and Infectious Disease Division, University of WashingtonSeattleUnited States
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, University of WashingtonSeattleUnited States
| | - Hans-Peter Kiem
- Department of Medicine, University of WashingtonSeattleUnited States
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Department of Pathology, University of WashingtonSeattleUnited States
| | - Joshua T Schiffer
- Vaccine and Infectious Disease Division, University of WashingtonSeattleUnited States
- Department of Medicine, University of WashingtonSeattleUnited States
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| |
Collapse
|
17
|
Rowell CER, Dobrovolny HM. Energy Requirements for Loss of Viral Infectivity. FOOD AND ENVIRONMENTAL VIROLOGY 2020; 12:281-294. [PMID: 32757142 PMCID: PMC7405386 DOI: 10.1007/s12560-020-09439-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/31/2020] [Indexed: 06/11/2023]
Abstract
Outside the host, viruses will eventually lose their ability to infect cells due to conformational changes that occur to proteins on the viral capsid. In order to undergo a conformational change, these proteins require energy to activate the chemical reaction that leads to the conformational change. In this study, data from the literature is used to calculate the energy required for viral inactivation for a variety of different viruses by means of the Arrhenius equation. We find that some viruses (rhinovirus, poliovirus, human immunodeficiency virus, Alkhumra hemorrhagic fever virus, and hepatitis A virus) have high inactivation energies, indicative of breaking of a chemical double bond. We also find that several viruses (respiratory syncytial virus, poliovirus, and norovirus) have nonlinear Arrhenius plots, suggesting that there is more than a single pathway for inactivation of these viruses.
Collapse
Affiliation(s)
- Caroline E R Rowell
- Department of Chemistry, Wingate University, Hendersonville, NC, USA
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, USA
| | - Hana M Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, USA.
| |
Collapse
|
18
|
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
|
19
|
Desikan R, Raja R, Dixit NM. Early exposure to broadly neutralizing antibodies may trigger a dynamical switch from progressive disease to lasting control of SHIV infection. PLoS Comput Biol 2020; 16:e1008064. [PMID: 32817614 PMCID: PMC7462315 DOI: 10.1371/journal.pcbi.1008064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/01/2020] [Accepted: 06/17/2020] [Indexed: 01/01/2023] Open
Abstract
Antiretroviral therapy (ART) for HIV-1 infection is life-long. Stopping therapy typically leads to the reignition of infection and progressive disease. In a major breakthrough, recent studies have shown that early initiation of ART can lead to sustained post-treatment control of viremia, raising hopes of long-term HIV-1 remission. ART, however, elicits post-treatment control in a small fraction of individuals treated. Strikingly, passive immunization with broadly neutralizing antibodies (bNAbs) of HIV-1 early in infection was found recently to elicit long-term control in a majority of SHIV-infected macaques, suggesting that HIV-1 remission may be more widely achievable. The mechanisms underlying the control elicited by bNAb therapy, however, remain unclear. Untreated infection typically leads to progressive disease. We hypothesized that viremic control represents an alternative but rarely realized outcome of the infection and that early bNAb therapy triggers a dynamical switch to this outcome. To test this hypothesis, we constructed a model of viral dynamics with bNAb therapy and applied it to analyse clinical data. The model fit quantitatively the complex longitudinal viral load data from macaques that achieved lasting control. The model predicted, consistently with our hypothesis, that the underlying system exhibited bistability, indicating two potential outcomes of infection. The first had high viremia, weak cytotoxic effector responses, and high effector exhaustion, marking progressive disease. The second had low viremia, strong effector responses, and low effector exhaustion, indicating lasting viremic control. Further, model predictions suggest that early bNAb therapy elicited lasting control via pleiotropic effects. bNAb therapy lowers viremia, which would also limit immune exhaustion. Simultaneously, it can improve effector stimulation via cross-presentation. Consequently, viremia may resurge post-therapy, but would encounter a primed effector population and eventually get controlled. ART suppresses viremia but does not enhance effector stimulation, explaining its limited ability to elicit post-treatment control relative to bNAb therapy. In a remarkable advance in HIV cure research, a recent study showed that 3 weekly doses of HIV-1 broadly neutralizing antibodies (bNAbs) soon after infection kept viral levels controlled for years in most macaques treated. If translated to humans, this bNAb therapy may elicit a functional cure, or long-term remission, of HIV-1 infection, eliminating the need for life-long antiretroviral therapy (ART). How early bNAb therapy works remains unknown. Here, we elucidate the mechanism using mathematical modeling and analysis of in vivo data. We predict that early bNAb therapy suppresses viremia, which reduces exhaustion of cytotoxic effector cells, and enhances antigen uptake and effector stimulation. Collectively, these effects drive infection to lasting control. Model predictions based on these effects fit in vivo data quantitatively. ART controls viremia but does not improve effector stimulation, explaining its weaker ability to induce lasting control post-treatment. Our findings may help improve strategies for achieving functional cure of HIV-1 infection.
Collapse
Affiliation(s)
- Rajat Desikan
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
- * E-mail: (RD); (NMD)
| | - Rubesh Raja
- 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: (RD); (NMD)
| |
Collapse
|
20
|
Rosenbloom DS, Zhao P, Sinha V. Initiation of Antiviral Treatment in SARS-CoV2: Modeling Viral Dynamics and Drug Properties. CPT Pharmacometrics Syst Pharmacol 2020; 9:481-483. [PMID: 32700405 PMCID: PMC7405045 DOI: 10.1002/psp4.12550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
- Daniel Scholes Rosenbloom
- Quantitative Pharmacology and Pharmacometrics, Pharmacokinetics, Pharmacodynamics, and Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Ping Zhao
- The Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Vikram Sinha
- Quantitative Pharmacology and Pharmacometrics, Pharmacokinetics, Pharmacodynamics, and Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
21
|
Alagaratnam J, Peters H, Francis K, Kay N, Gilleece Y, Finnerty FP, Grimes RE, Parry S, Portman M, Wait BC, Shah R, Roedling S, Hawkins DA, Chitty S, Sarner L, Marcus R, Hartley A, Nori AV, Rosenvinge M, Taylor GP. An observational study of initial HIV RNA decay following initiation of combination antiretroviral treatment during pregnancy. AIDS Res Ther 2020; 17:41. [PMID: 32660502 PMCID: PMC7359473 DOI: 10.1186/s12981-020-00297-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/04/2020] [Indexed: 01/29/2023] Open
Abstract
Background In pregnancy, reduction of HIV plasma viral load (pVL) for the prevention of vertical transmission is time-constrained. The study primary objective is to investigate factors associated with faster initial HIV RNA half-life decay when combination antiretroviral treatment (cART) is initiated in pregnancy. Methods This was a multicentre, retrospective, observational study, conducted in south England, United Kingdom, between August 2001 and February 2018. Data were extracted from case notes of eligible women initiating cART during the index pregnancy. Anonymised data were collated and analysed centrally. Regression analyses were conducted to determine factors associated with faster HIV RNA half-life decay in the first 14 days after commencing cART (first-phase), and with achieving an undetectable maternal pVL by 36 weeks’ gestation. We then assessed whether HIV- and obstetric- related parameters differed by antiretroviral third agent class and whether the proportions of women with undetectable pVL at 36 weeks’ gestation and at delivery differed by antiretroviral third agent class. Results Baseline pVL was the only independent factor associated with faster first-phase HIV RNA half-life decay on commencing cART. Lower pVL on day 14 after starting cART was associated with an increased likelihood of achieving an undetectable pVL by 36 weeks’ gestation. Integrase inhibitor-based cART was associated with a faster first-phase HIV RNA half-life decay on commencing cART. Overall, 73% and 85% of women had an undetectable pVL at 36 weeks’ gestation and at delivery respectively, with no significant difference by antiretroviral third agent class. Conclusions Only high baseline pVL independently contributed to a faster rate of first-phase viral half-life decay. pVL at 14 days after initiating cART allows early identification of treatment failure. In the first 14 days after initiating cART in pregnancy, integrase inhibitor-based cART reduced maternal pVL faster than protease inhibitor- and non-nucleoside reverse transcriptase-based cART. While our study findings support INSTI use when initiated in pregnancy especially when initiated at later gestations and in those with higher baseline pVL, other non-INSTI based cART with more data on safety in pregnancy also performed well.
Collapse
|
22
|
Pinzone MR, Bertuccio MP, VanBelzen DJ, Zurakowski R, O'Doherty U. Next-Generation Sequencing in a Direct Model of HIV Infection Reveals Important Parallels to and Differences from In Vivo Reservoir Dynamics. J Virol 2020; 94:e01900-19. [PMID: 32051279 PMCID: PMC7163122 DOI: 10.1128/jvi.01900-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Next-generation sequencing (NGS) represents a powerful tool to unravel the genetic make-up of the HIV reservoir, but limited data exist on its use in vitro Moreover, most NGS studies do not separate integrated from unintegrated DNA, even though selection pressures on these two forms should be distinct. We reasoned we could use NGS to compare the infection of resting and activated CD4 T cells in vitro to address how the metabolic state affects reservoir formation and dynamics. To address these questions, we obtained HIV sequences 2, 4, and 8 days after NL4-3 infection of metabolically activated and quiescent CD4 T cells (cultured with 2 ng/ml interleukin-7). We compared the composition of integrated and total HIV DNA by isolating integrated HIV DNA using pulsed-field electrophoresis before performing sequencing. After a single-round infection, the majority of integrated HIV DNA was intact in both resting and activated T cells. The decay of integrated intact proviruses was rapid and similar in both quiescent and activated T cells. Defective forms accumulated relative to intact ones analogously to what is observed in vivo Massively deleted viral sequences formed more frequently in resting cells, likely due to lower deoxynucleoside triphosphate (dNTP) levels and the presence of multiple restriction factors. To our surprise, the majority of these deleted sequences did not integrate into the human genome. The use of NGS to study reservoir dynamics in vitro provides a model that recapitulates important aspects of reservoir dynamics. Moreover, separating integrated from unintegrated HIV DNA is important in some clinical settings to properly study selection pressures.IMPORTANCE The major implication of our work is that the decay of intact proviruses in vitro is extremely rapid, perhaps as a result of enhanced expression. Gaining a better understanding of why intact proviruses decay faster in vitro might help the field identify strategies to purge the reservoir in vivo When used wisely, in vitro models are a powerful tool to study the selective pressures shaping the viral landscape. Our finding that massively deleted sequences rarely succeed in integrating has several ramifications. It demonstrates that the total HIV DNA can differ substantially in character from the integrated HIV DNA under certain circumstances. The presence of unintegrated HIV DNA has the potential to obscure selection pressures and confound the interpretation of clinical studies, especially in the case of trials involving treatment interruptions.
Collapse
Affiliation(s)
- Marilia Rita Pinzone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria Paola Bertuccio
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - D Jake VanBelzen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, USA
| | - Ryan Zurakowski
- Delaware Biotechnology Institute, University of Delaware, Newark, Delaware, USA
| | - Una O'Doherty
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Morris SE, Dziobek-Garrett L, Yates AJ, Collaboration With The Epiical Consortium I. ushr: Understanding suppression of HIV in R. BMC Bioinformatics 2020; 21:52. [PMID: 32046642 PMCID: PMC7014720 DOI: 10.1186/s12859-020-3389-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/28/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND HIV/AIDS is responsible for the deaths of one million people every year. Although mathematical modeling has provided many insights into the dynamics of HIV infection, there is still a lack of accessible tools for researchers unfamiliar with modeling techniques to apply them to their own clinical data. RESULTS Here we present ushr, a free and open-source R package that models the decline of HIV during antiretroviral treatment (ART) using a popular mathematical framework. ushr can be applied to longitudinal data of viral load measurements, and provides processing tools to prepare it for computational analysis. By mathematically fitting the data, important biological parameters can then be estimated, including the lifespans of short and long-lived infected cells, and the time to reach viral suppression below a defined detection threshold. The package also provides visualization and summary tools for fast assessment of model results. CONCLUSIONS ushr enables researchers without a strong mathematical or computational background to model the dynamics of HIV using longitudinal clinical data. Increasing accessibility to such methods may facilitate quantitative analysis across a broader range of independent studies, so that greater insights on HIV infection and treatment dynamics may be gained.
Collapse
Affiliation(s)
- Sinead E Morris
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| | - Luise Dziobek-Garrett
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | | |
Collapse
|
24
|
Engelman AN. Multifaceted HIV integrase functionalities and therapeutic strategies for their inhibition. J Biol Chem 2019; 294:15137-15157. [PMID: 31467082 DOI: 10.1074/jbc.rev119.006901] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antiretroviral inhibitors that are used to manage HIV infection/AIDS predominantly target three enzymes required for virus replication: reverse transcriptase, protease, and integrase. Although integrase inhibitors were the last among this group to be approved for treating people living with HIV, they have since risen to the forefront of treatment options. Integrase strand transfer inhibitors (INSTIs) are now recommended components of frontline and drug-switch antiretroviral therapy formulations. Integrase catalyzes two successive magnesium-dependent polynucleotidyl transferase reactions, 3' processing and strand transfer, and INSTIs tightly bind the divalent metal ions and viral DNA end after 3' processing, displacing from the integrase active site the DNA 3'-hydroxyl group that is required for strand transfer activity. Although second-generation INSTIs present higher barriers to the development of viral drug resistance than first-generation compounds, the mechanisms underlying these superior barrier profiles are incompletely understood. A separate class of HIV-1 integrase inhibitors, the allosteric integrase inhibitors (ALLINIs), engage integrase distal from the enzyme active site, namely at the binding site for the cellular cofactor lens epithelium-derived growth factor (LEDGF)/p75 that helps to guide integration into host genes. ALLINIs inhibit HIV-1 replication by inducing integrase hypermultimerization, which precludes integrase binding to genomic RNA and perturbs the morphogenesis of new viral particles. Although not yet approved for human use, ALLINIs provide important probes that can be used to investigate the link between HIV-1 integrase and viral particle morphogenesis. Herein, I review the mechanisms of retroviral integration as well as the promises and challenges of using integrase inhibitors for HIV/AIDS management.
Collapse
Affiliation(s)
- Alan N Engelman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
25
|
Goonetilleke N, Clutton G, Swanstrom R, Joseph SB. Blocking Formation of the Stable HIV Reservoir: A New Perspective for HIV-1 Cure. Front Immunol 2019; 10:1966. [PMID: 31507594 PMCID: PMC6714000 DOI: 10.3389/fimmu.2019.01966] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022] Open
Abstract
Recent studies demonstrate that the stable HIV-1 reservoir in resting CD4+ T cells is mostly formed from viruses circulating when combination antiretroviral therapy (ART) is initiated. Here we explore the immunological basis for these observations. Untreated HIV-1 infection is characterized by a progressive depletion of memory CD4+ T cells which mostly express CD127, the α chain of the IL-7 receptor (IL-7R). Depletion results from both direct infection and bystander loss of memory CD4+ T cells in part attributed to dysregulated IL-7/IL-7R signaling. While IL-7/IL7R signaling is not essential for the generation of effector CD4+ T cells from naïve cells, it is essential for the further transition of effectors to memory CD4+ T cells and their subsequent homeostatic maintenance. HIV-1 infection therefore limits the transition of CD4+ T cells from an effector to long-lived memory state. With the onset of ART, virus load (VL) levels rapidly decrease and the frequency of CD127+ CD4+ memory T cells increases, indicating restoration of effector to memory transition in CD4+ T cells. Collectively these data suggest that following ART initiation, HIV-1 infected effector CD4+ T cells transition to long-lived, CD127+ CD4+ T cells forming the majority of the stable HIV-1 reservoir. We propose that combining ART initiation with inhibition of IL-7/IL-7R signaling to block CD4+ T cell memory formation will limit the generation of long-lived HIV-infected CD4+ T cells and reduce the overall size of the stable HIV-1 reservoir.
Collapse
Affiliation(s)
- Nilu Goonetilleke
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- UNC HIV-1 Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Genevieve Clutton
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- UNC HIV-1 Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ron Swanstrom
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sarah B. Joseph
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
26
|
Veldsman KA, Janse van Rensburg A, Isaacs S, Naidoo S, Laughton B, Lombard C, Cotton MF, Mellors JW, van Zyl GU. HIV-1 DNA decay is faster in children who initiate ART shortly after birth than later. J Int AIDS Soc 2019; 22:e25368. [PMID: 31441231 PMCID: PMC6868516 DOI: 10.1002/jia2.25368] [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: 02/05/2019] [Accepted: 07/15/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION There is limited data in children on whether persistence of HIV-1 infected cells is affected by age at initiating antiretroviral therapy (ART), its duration or any subsequent ART interruption. We therefore investigated the effects of both age of ART initiation and duration of ART interruption on HIV-1 DNA decay in children. METHODS We investigated HIV-1 DNA decay in three groups of children on ART: Group-1 (n = 7) started uninterrupted ART within eight days of life; Group-2 (n = 8) started uninterrupted ART at a median of five months of age; and Group-3 (n = 23) started ART at a median age of 1.8 months for either 40 or 96 weeks, then interrupted ART (median of seven months), and restarted ART based on CD4 count and clinical criteria. Total HIV-1 DNA was assayed using a sensitive HIV-1 subtype C-adapted quantitative PCR for integrase. The duration of ART was square root transformed to fit the observed slowing of HIV-1 DNA decay rate. For each group, point estimates for decay rates were determined after six months of continuous suppressive ART in groups 1 and 2 or six months after restarting ART in Group-3. Groups-2 and 3 were combined using a mixed effect regression model to investigate covariates of HIV-1 DNA decay rate. RESULTS AND DISCUSSION At six months of continuous suppressive ART, the HIV-1 DNA t½ (95% CI) was shorter in Group-1 (n = 7): 2.7 months (2.1 to 3.8), than 9.2 months (7.4 to 12.1) in Group-2 (n = 8); and 9.6 months (7.6 to 12.6) in Group-3 (n = 23) (p < 0.01). In multivariable analyses, HIV-1 DNA before treatment (p < 0.001) and the change in HIV-1 DNA during interruption (p < 0.01) were independent predictors of slower HIV-1 DNA decay. CONCLUSIONS These data suggest that ART initiation within the first week of life can reduce the persistence of long-lived infected cells. Delaying ART is associated with slower decay of infected cells.
Collapse
Affiliation(s)
- Kirsten A Veldsman
- Division of Medical VirologyStellenbosch UniversityFaculty of Medicine and Health SciencesCape TownSouth Africa
| | - Anita Janse van Rensburg
- Department Pediatrics and Child HealthTygerberg Children's Hospital and Family Clinical Research UnitStellenbosch UniversityCape TownSouth Africa
| | - Shahieda Isaacs
- Division of Medical VirologyStellenbosch UniversityFaculty of Medicine and Health SciencesCape TownSouth Africa
| | - Shalena Naidoo
- Division of Medical VirologyStellenbosch UniversityFaculty of Medicine and Health SciencesCape TownSouth Africa
| | - Barbara Laughton
- Department Pediatrics and Child HealthTygerberg Children's Hospital and Family Clinical Research UnitStellenbosch UniversityCape TownSouth Africa
| | - Carl Lombard
- Division of Epidemiology and BiostatisticsDepartment of Global HealthFaculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
- Biostatistics UnitSouth African Medical Research CouncilCape TownSouth Africa
| | - Mark F Cotton
- Department Pediatrics and Child HealthTygerberg Children's Hospital and Family Clinical Research UnitStellenbosch UniversityCape TownSouth Africa
| | - John W Mellors
- Division of Infectious DiseasesDepartment of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Gert U van Zyl
- Division of Medical VirologyStellenbosch UniversityFaculty of Medicine and Health SciencesCape TownSouth Africa
- National Health Laboratory ServiceCape TownSouth Africa
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW A number studies are currently underway to develop new drugs aimed at reducing the HIV reservoir or achieving ART-free control of HIV infection. Many markers of HIV reservoirs have been proposed, each one having a different meaning. Total HIV DNA dynamics during the course of HIV infection and its predictive value are now well known. This marker allowed to estimate the size of HIV reservoir at different stages of HIV infection in blood, cell subsets and tissues. Therefore, the purpose of this review is timely and relevant, with the objective to discuss how total HIV DNA might be helpful in the clinical settings. RECENT FINDINGS Among the markers, it appears that HIV DNA is the most well studied, and recent articles confirmed that this marker is easy to use and is precise, specific, practical, robust and reproducible. All these characteristics correspond to what is expected from a helpful clinical marker. SUMMARY HIV DNA level could be considered as a global marker, and it is usually included in current clinical studies to describe the persistence and dynamics of the HIV reservoir, mainly in treated patients. HIV DNA might be helpful in designing clinical trials and personalized medication for HIV patients in the future.
Collapse
|
28
|
Gantner P, Sylla B, Morand-Joubert L, Frange P, Lacombe K, Khuong MA, Duvivier C, Launay O, Karmochkine M, Arvieux C, Ménard A, Piroth L, Canestri A, Trias D, Peytavin G, Landman R, Ghosn J. "Real life" use of raltegravir during pregnancy in France: The Coferal-IMEA048 cohort study. PLoS One 2019; 14:e0216010. [PMID: 31017957 PMCID: PMC6481866 DOI: 10.1371/journal.pone.0216010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/11/2019] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Limited "real life" data on raltegravir (RAL) use during pregnancy are available. Thus, we aimed at describing effectiveness and safety of RAL-based combined antiretroviral therapy (cART) in this setting. METHODS HIV-1-infected women receiving RAL during pregnancy between 2008 and 2014 in ten French centers were retrospectively analysed for: (1) proportion of women receiving RAL anytime during pregnancy who achieved a plasma HIV-RNA (pVL) < 50 copies/mL at delivery, and (2) description of demographics, immuno-virological parameters and safety in women and new-borns. RESULTS We included 94 women (median age, 33 years) of which 85% originated from Sub-Saharan Africa and 16% did not have regular health insurance coverage. Sixteen women were cART-naïve (median HIV diagnosis at 30 weeks of gestation), whereas 78 were already on cART before pregnancy (40% with pVL < 50 copies/mL). RAL was initiated before pregnancy (n = 33), during the second trimester (n = 11) and the third trimester of pregnancy (n = 50). No RAL discontinuations due to adverse events were observed. Overall, at the time of delivery, pVL was < 50 copies/mL in 70% and < 400 copies/mL in 84% of women. Specifically, pVL at delivery was < 50 copies/mL in 82%, 55% and 56% of cases when RAL was started before pregnancy, during the second or third trimester of pregnancy, respectively. Median term was 38 weeks of gestation, no defect was reported and all new-borns were HIV non-infected at Month 6. CONCLUSIONS RAL appears safe and effective in this "real-life" study. No defect and no HIV transmission was reported in new-borns.
Collapse
Affiliation(s)
- Pierre Gantner
- Hôpitaux Universitaires de Strasbourg, Laboratoire de Virologie, Strasbourg, France
| | - Babacar Sylla
- IMEA, CHU Bichat Claude Bernard, Paris, France Paris, France
| | - Laurence Morand-Joubert
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136), AP-HP, Laboratoire de Virologie, Hôpital Saint-Antoine, Paris, France
| | - Pierre Frange
- APHP, Hopital Necker Enfants malades, Laboratoire de Microbiologie clinique, Paris, France
- EHU 7328, Institut Imagine, Université Paris Descartes, Paris, France
| | - Karine Lacombe
- Inserm UMR-S1136, IPLESP, AP-HP, Hôpital Saint Antoine, Department of Infectious Diseases, Paris, France
| | - Marie-Aude Khuong
- Hôpital Delafontaine, Department of Infectious Diseases, Saint Denis, France
| | - Claudine Duvivier
- APHP, Hopital Necker Enfants Malades, Department of Infectious Diseases, Centre d’Infectiologie Necker – Pasteur, IHU Imagine, Paris, France
| | - Odile Launay
- Université Paris Descartes, APHP, CIC Cochin Pasteur, Paris, France
| | - Marina Karmochkine
- APHP, Hopital Européen Georges Pompidou, Department of Clinical Immunology, Paris, France
| | | | - Amélie Ménard
- Institut hospitalo-universitaire (IHU) Méditerranée infection, Marseille, France
| | - Lionel Piroth
- Département d’Infectiologie, CHU Dijon, Dijon, France
| | - Ana Canestri
- APHP, Hôpital Tenon, Maladies Infectieuses, Paris, France
| | | | - Gilles Peytavin
- APHP, Hopital Bichat Claude Bernard, Department of Pharmacology-Toxicology, Paris, France
- INSERM IAME UMR-S 1137, Université Paris Diderot, Paris, France
| | - Roland Landman
- IMEA, CHU Bichat Claude Bernard, Paris, France Paris, France
- INSERM IAME UMR-S 1137, Université Paris Diderot, Paris, France
- APHP, Hopital Bichat Claude Bernard, Department of Infectious Diseases, Paris, France
| | - Jade Ghosn
- INSERM IAME UMR-S 1137, Université Paris Diderot, Paris, France
- APHP, Hopital Bichat Claude Bernard, Department of Infectious Diseases, Paris, France
| | | |
Collapse
|
29
|
Trémeaux P, Lenfant T, Boufassa F, Essat A, Mélard A, Gousset M, Delelis O, Viard JP, Bary M, Goujard C, Rouzioux C, Meyer L, Avettand-Fenoel V. Increasing contribution of integrated forms to total HIV DNA in blood during HIV disease progression from primary infection. EBioMedicine 2019; 41:455-464. [PMID: 30803934 PMCID: PMC6442355 DOI: 10.1016/j.ebiom.2019.02.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/25/2019] [Accepted: 02/06/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND In the current context of research on HIV reservoirs, offering new insights into the persistence of HIV DNA in infected cells, which prevents viral eradication, may aid in identifying cure strategies. This study aimed to describe the establishment of stable integrated forms among total HIV DNA during primary infection (PHI) and their dynamics during the natural history of infection. METHODS Total and integrated HIV DNA were quantified in blood from 74 PHI patients and 97 recent seroconverters (<12 months following infection, "progression cohort"). The evolution of both markers over six years was modelled (mixed-effect linear models). Their predictive values for disease progression were studied (Cox models). FINDINGS For most patients during PHI, stable integrated forms were a minority among total HIV DNA (median: 12%) and became predominant thereafter (median at AIDS stage: 100%). Both total and integrated HIV DNA increased over a six-year period. Patients from the progression cohort who reached clinical AIDS during follow-up (n = 34) exhibited higher total and integrated HIV DNA levels at seroconversion and a higher percentage of integrated forms than did slower progressors (n = 63) (median: 100% vs 44%). The integrated HIV DNA load was strongly associated with the risk of developing AIDS (aRR = 2.63, p = 0.002). INTERPRETATION The profile of "rapid" or "slower" progression in the natural history of HIV infection appears to be determined early in the course of HIV infection. The strong predominance of unstable unintegrated forms in PHI may explain the great benefit of this early treatment, which induces a sharp decrease in total HIV DNA. FUND: French National Agency for Research on AIDS and Viral Hepatitis.
Collapse
Affiliation(s)
- Pauline Trémeaux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; AP-HP, Laboratoire de Virologie, Hôpital Cochin, Paris, France
| | - Tiphaine Lenfant
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Faroudy Boufassa
- Université Paris Sud, Université Paris Saclay, INSERM CESP U1018, le Kremlin-Bicêtre, France
| | - Asma Essat
- Université Paris Sud, Université Paris Saclay, INSERM CESP U1018, le Kremlin-Bicêtre, France; AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Adeline Mélard
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France
| | - Marine Gousset
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France
| | - Olivier Delelis
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre National de la Recherche Scientifique UMR8113, Cachan, France
| | - Jean-Paul Viard
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; AP-HP, Centre de diagnostic et thérapeutique, Hôtel-Dieu, Paris, France
| | - Marc Bary
- AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Cécile Goujard
- Université Paris Sud, Université Paris Saclay, INSERM CESP U1018, le Kremlin-Bicêtre, France; AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christine Rouzioux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Laurence Meyer
- Université Paris Sud, Université Paris Saclay, INSERM CESP U1018, le Kremlin-Bicêtre, France; AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Véronique Avettand-Fenoel
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; AP-HP, Laboratoire de Microbiologie clinique, CHU Necker-Enfants Malades, Paris, France.
| |
Collapse
|
30
|
Pinzone MR, VanBelzen DJ, Weissman S, Bertuccio MP, Cannon L, Venanzi-Rullo E, Migueles S, Jones RB, Mota T, Joseph SB, Groen K, Pasternak AO, Hwang WT, Sherman B, Vourekas A, Nunnari G, O'Doherty U. Longitudinal HIV sequencing reveals reservoir expression leading to decay which is obscured by clonal expansion. Nat Commun 2019; 10:728. [PMID: 30760706 PMCID: PMC6374386 DOI: 10.1038/s41467-019-08431-7] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 12/31/2018] [Indexed: 01/17/2023] Open
Abstract
After initiating antiretroviral therapy (ART), a rapid decline in HIV viral load is followed by a long period of undetectable viremia. Viral outgrowth assay suggests the reservoir continues to decline slowly. Here, we use full-length sequencing to longitudinally study the proviral landscape of four subjects on ART to investigate the selective pressures influencing the dynamics of the treatment-resistant HIV reservoir. We find intact and defective proviruses that contain genetic elements favoring efficient protein expression decrease over time. Moreover, proviruses that lack these genetic elements, yet contain strong donor splice sequences, increase relatively to other defective proviruses, especially among clones. Our work suggests that HIV expression occurs to a significant extent during ART and results in HIV clearance, but this is obscured by the expansion of proviral clones. Paradoxically, clonal expansion may also be enhanced by HIV expression that leads to splicing between HIV donor splice sites and downstream human exons.
Collapse
Affiliation(s)
- Marilia Rita Pinzone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - D Jake VanBelzen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Molecular Biosciences, Northwestern University, Evanston, 60201, IL, USA
| | - Sam Weissman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Maria Paola Bertuccio
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - LaMont Cannon
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Emmanuele Venanzi-Rullo
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Messina, 98124, Italy
| | - Stephen Migueles
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, 20892, MD, USA
| | - R Brad Jones
- Infectious Disease Division, Weill Cornell Medical College, New York, 10065, NY, USA
| | - Talia Mota
- Infectious Disease Division, Weill Cornell Medical College, New York, 10065, NY, USA
| | - Sarah B Joseph
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
| | - Kevin Groen
- Laboratory of Experimental Virology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, The Netherlands
| | - Alexander O Pasternak
- Laboratory of Experimental Virology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, The Netherlands
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Brad Sherman
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratories for Cancer Research, Leidos Biomedical Research Inc., supporting the Division of Clinical Research, NIAID, Frederick, 21702, MD, USA
| | - Anastasios Vourekas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Messina, 98124, Italy
| | - Una O'Doherty
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA.
| |
Collapse
|
31
|
A majority of HIV persistence during antiretroviral therapy is due to infected cell proliferation. Nat Commun 2018; 9:4811. [PMID: 30446650 PMCID: PMC6240116 DOI: 10.1038/s41467-018-06843-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
Antiretroviral therapy (ART) suppresses viral replication in people living with HIV. Yet, infected cells persist for decades on ART and viremia returns if ART is stopped. Persistence has been attributed to viral replication in an ART sanctuary and long-lived and/or proliferating latently infected cells. Using ecological methods and existing data, we infer that >99% of infected cells are members of clonal populations after one year of ART. We reconcile our results with observations from the first months of ART, demonstrating mathematically how a fossil record of historic HIV replication permits observed viral evolution even while most new infected cells arise from proliferation. Together, our results imply cellular proliferation generates a majority of infected cells during ART. Therefore, reducing proliferation could decrease the size of the HIV reservoir and help achieve a functional cure. HIV infected cells persist for decades in patients under ART, but the mechanisms responsible remain unclear. Here, Reeves et al. use modeling approaches adapted from ecology to show that cellular proliferation, rather than viral replication, generates a majority of infected cells during ART.
Collapse
|
32
|
Ke R, Conway JM, Margolis DM, Perelson AS. Determinants of the efficacy of HIV latency-reversing agents and implications for drug and treatment design. JCI Insight 2018; 3:123052. [PMID: 30333308 DOI: 10.1172/jci.insight.123052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/30/2018] [Indexed: 11/17/2022] Open
Abstract
HIV eradication studies have focused on developing latency-reversing agents (LRAs). However, it is not understood how the rate of latent reservoir reduction is affected by different steps in the process of latency reversal. Furthermore, as current LRAs are host-directed, LRA treatment is likely to be intermittent to avoid host toxicities. Few careful studies of the serial effects of pulsatile LRA treatment have yet been done. This lack of clarity makes it difficult to evaluate the efficacy of candidate LRAs or predict long-term treatment outcomes. We constructed a mathematical model that describes the dynamics of latently infected cells under LRA treatment. Model analysis showed that, in addition to increasing the immune recognition and clearance of infected cells, the duration of HIV antigen expression (i.e., the period of vulnerability) plays an important role in determining the efficacy of LRAs, especially if effective clearance is achieved. Patients may benefit from pulsatile LRA exposures compared with continuous LRA exposures if the period of vulnerability is long and the clearance rate is high, both in the presence and absence of an LRA. Overall, the model framework serves as a useful tool to evaluate the efficacy and the rational design of LRAs and combination strategies.
Collapse
Affiliation(s)
- Ruian Ke
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA.,Theoretical Biology and Biophysics Group, MS-K710, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Jessica M Conway
- Department of Mathematics and Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, USA
| | - David M Margolis
- University of North Carolina (UNC) HIV Cure Center, UNC Institute of Global Health and Infectious Diseases.,Departments of Medicine, Microbiology and Immunology, UNC Chapel Hill School of Medicine, and.,Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, MS-K710, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| |
Collapse
|
33
|
Jacobson K, Ogbuagu O. Integrase inhibitor-based regimens result in more rapid virologic suppression rates among treatment-naïve human immunodeficiency virus-infected patients compared to non-nucleoside and protease inhibitor-based regimens in a real-world clinical setting: A retrospective cohort study. Medicine (Baltimore) 2018; 97:e13016. [PMID: 30412140 PMCID: PMC6221636 DOI: 10.1097/md.0000000000013016] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The integrase strand transfer inhibitor (INSTI) class of antiretroviral therapy (ART) may result in faster time to virologic suppression compared with regimens that contain protease inhibitors (PIs) or non-nucleoside reverse transcriptase inhibitors (NNRTIs). However, differences in time to achieve virologic suppression are not well-defined in routine clinical settings with contemporary antiretroviral agents.Study was a retrospective single-center study of treatment-naïve human immunodeficiency virus (HIV) patients initiating ART between 2013 and 2016. Among patients on different ART regimen types, we compared rates of and median time to virologic suppression [viral load (VL) <50 copies/mL].A total of 155 patients-45 (29%) female and 110 (71%) male-met study inclusion criteria. Median age was 42 years (interquartile range 31-52), and median baseline CD4 count was 288 cells/μL and VL was 60,000 copies/mL. Seventy-one (46%) initiated an INSTI-based regimen, 58 (37%) were on NNRTI-based regimens, and 26 (17%) on PI-based regimens. In total, 112 (72%) patients achieved virologic suppression at 12 months. Patients on INSTI-based regimens were more likely to achieve virologic suppression by 3, 6, and 12 months (P < .01), and had lower median time to suppression (60 vs 137 days on NNRTI-based regimens and 147 days on PI-based regimens, P < .01).Patients on INSTI-based ART regimens in a real-world setting experienced higher rates of virologic suppression and shorter time from ART initiation to virologic suppression. For HIV patients on INSTI-based ART regimens, virologic failure should be suspected in those with VLs >50 copies/mL before the current recommendation of 48 weeks.
Collapse
Affiliation(s)
| | - Onyema Ogbuagu
- Yale AIDS Program, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
34
|
Cardozo EF, Apetrei C, Pandrea I, Ribeiro RM. The dynamics of simian immunodeficiency virus after depletion of CD8+ cells. Immunol Rev 2018; 285:26-37. [PMID: 30129200 PMCID: PMC6352983 DOI: 10.1111/imr.12691] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human immunodeficiency virus infection is still one of the most important causes of morbidity and mortality in the world, with a disproportionate human and economic burden especially in poorer countries. Despite many years of intense research, an aspect that still is not well understood is what (immune) mechanisms control the viral load during the prolonged asymptomatic stage of infection. Because CD8+ T cells have been implicated in this control by multiple lines of evidence, there has been a focus on understanding the potential mechanisms of action of this immune effector population. One type of experiment used to this end has been depleting these cells with monoclonal antibodies in the simian immunodeficiency virus-macaque model and then studying the effect of that depletion on the viral dynamics. Here we review what these experiments have told us. We emphasize modeling studies to interpret the changes in viral load observed in these experiments, including discussion of alternative models, assumptions and interpretations, as well as potential future experiments.
Collapse
Affiliation(s)
- Erwing Fabian Cardozo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
- Laboratorio de Biomatematica, Faculdade de Medicina da Universidade de Lisboa, Portugal
| |
Collapse
|
35
|
Hill AL, Rosenbloom DIS, Nowak MA, Siliciano RF. Insight into treatment of HIV infection from viral dynamics models. Immunol Rev 2018; 285:9-25. [PMID: 30129208 PMCID: PMC6155466 DOI: 10.1111/imr.12698] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The odds of living a long and healthy life with HIV infection have dramatically improved with the advent of combination antiretroviral therapy. Along with the early development and clinical trials of these drugs, and new field of research emerged called viral dynamics, which uses mathematical models to interpret and predict the time-course of viral levels during infection and how they are altered by treatment. In this review, we summarize the contributions that virus dynamics models have made to understanding the pathophysiology of infection and to designing effective therapies. This includes studies of the multiphasic decay of viral load when antiretroviral therapy is given, the evolution of drug resistance, the long-term persistence latently infected cells, and the rebound of viremia when drugs are stopped. We additionally discuss new work applying viral dynamics models to new classes of investigational treatment for HIV, including latency-reversing agents and immunotherapy.
Collapse
Affiliation(s)
- Alison L. Hill
- Program for Evolutionary DynamicsHarvard UniversityCambridgeMassachusetts
| | - Daniel I. S. Rosenbloom
- Department of PharmacokineticsPharmacodynamics, & Drug MetabolismMerck Research LaboratoriesKenilworthNew Jersey
| | - Martin A. Nowak
- Program for Evolutionary DynamicsHarvard UniversityCambridgeMassachusetts
| | - Robert F. Siliciano
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMaryland
- Howard Hughes Medical InstituteBaltimoreMaryland
| |
Collapse
|
36
|
Exploring an alternative explanation for the second phase of viral decay: Infection of short-lived cells in a drug-limited compartment during HAART. PLoS One 2018; 13:e0198090. [PMID: 30016329 PMCID: PMC6049925 DOI: 10.1371/journal.pone.0198090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/14/2018] [Indexed: 11/19/2022] Open
Abstract
Most HIV-infected patients who initiate combination antiretroviral therapy experience a viral load decline in several phases. These phases are characterized by different rates of viral load decay that decrease when transitioning from one phase to the next. There is no consensus as to the origin of these phases. One hypothesis put forward is that short- and long-lived infected cells are responsible for the first and second phases of decay, respectively. However, significant differences in drug concentrations are observed in monocytes from various tissues, suggesting the first two phases of decay in viral loads could instead be attributed to short-lived cells being differently exposed to drugs. Compared to a well-exposed compartment, new cell infection can be expected in a compartment with limited drug exposure, thus leading to a slower viral load decay with potential virologic failure and drug resistance. In the current study, the latter hypothesis was investigated using a model of viral kinetics. Empirical datasets were involved in model elaboration and parameter estimation. In particular, susceptibility assay data was used for an in vitro to in vivo extrapolation based on the expected drug concentrations inside physiological compartments. Results from numerical experiments of the short-term evolution of viral loads can reproduce the first two phases of viral decay when allowing new short-lived cell infections in an unidentified drug-limited compartment. Model long-term predictions are however less consistent with clinical observations. For the hypothesis to hold, efavirenz, tenofovir and emtricitabine drug exposure in the drug-limited compartment would have to be very low compared to exposure in peripheral blood. This would lead to significant long-term viral growth and the frequent development of resistant strains, a prediction not supported by clinical observations. This suggests that the existence of a drug-limited anatomical compartment is unlikely, by itself, to explain the second phase of viral load decay.
Collapse
|
37
|
Zitzmann C, Kaderali L. Mathematical Analysis of Viral Replication Dynamics and Antiviral Treatment Strategies: From Basic Models to Age-Based Multi-Scale Modeling. Front Microbiol 2018; 9:1546. [PMID: 30050523 PMCID: PMC6050366 DOI: 10.3389/fmicb.2018.01546] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/21/2018] [Indexed: 12/14/2022] Open
Abstract
Viral infectious diseases are a global health concern, as is evident by recent outbreaks of the middle east respiratory syndrome, Ebola virus disease, and re-emerging zika, dengue, and chikungunya fevers. Viral epidemics are a socio-economic burden that causes short- and long-term costs for disease diagnosis and treatment as well as a loss in productivity by absenteeism. These outbreaks and their socio-economic costs underline the necessity for a precise analysis of virus-host interactions, which would help to understand disease mechanisms and to develop therapeutic interventions. The combination of quantitative measurements and dynamic mathematical modeling has increased our understanding of the within-host infection dynamics and has led to important insights into viral pathogenesis, transmission, and disease progression. Furthermore, virus-host models helped to identify drug targets, to predict the treatment duration to achieve cure, and to reduce treatment costs. In this article, we review important achievements made by mathematical modeling of viral kinetics on the extracellular, intracellular, and multi-scale level for Human Immunodeficiency Virus, Hepatitis C Virus, Influenza A Virus, Ebola Virus, Dengue Virus, and Zika Virus. Herein, we focus on basic mathematical models on the population scale (so-called target cell-limited models), detailed models regarding the most important steps in the viral life cycle, and the combination of both. For this purpose, we review how mathematical modeling of viral dynamics helped to understand the virus-host interactions and disease progression or clearance. Additionally, we review different types and effects of therapeutic strategies and how mathematical modeling has been used to predict new treatment regimens.
Collapse
Affiliation(s)
- Carolin Zitzmann
- Institute of Bioinformatics and Center for Functional Genomics of Microbes, University Medicine Greifswald, Greifswald, Germany
| | - Lars Kaderali
- Institute of Bioinformatics and Center for Functional Genomics of Microbes, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
38
|
Dynamics of Simian Immunodeficiency Virus Two-Long-Terminal-Repeat Circles in the Presence and Absence of CD8 + Cells. J Virol 2018; 92:JVI.02100-17. [PMID: 29643246 DOI: 10.1128/jvi.02100-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 04/08/2018] [Indexed: 12/28/2022] Open
Abstract
CD8+ cells play a key role in human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection, but their specific mechanism(s) of action in controlling the virus is unclear. Two-long-terminal-repeat (2-LTR) circles are extrachromosomal products generated upon failed integration of HIV/SIV. To understand the specific effects of CD8+ cells on infected cells, we analyzed the dynamics of 2-LTR circles in SIVmac251-infected rhesus macaques (RMs) treated with an integrase inhibitor (INT). Twenty RMs underwent CD8+ cell depletion and received raltegravir (RAL) monotherapy or a combination of both. Blood, lymph nodes (LNs), and gut biopsy specimens were routinely sampled. Plasma viral loads (pVLs) and 2-LTR circles from peripheral blood mononuclear cells (PBMCs) and LN lymphocytes were measured with quantitative reverse transcription-PCR (qRT-PCR). In the CD8 depletion group, an ∼1-log increase in pVLs and a slow increase in PBMC 2-LTRs occurred following depletion. In the INT group, a strong decline in pVLs upon treatment initiation and no change in 2-LTR levels were observed. In the INT and CD8+ cell depletion group, an increase in pVLs following CD8 depletion similar to that in the CD8 depletion group was observed, with a modest decline following INT initiation, and 2-LTR circles significantly increased in PBMCs and LNs. Analyzing the 2-LTR data across all treatment groups with a mathematical model indicates that the data best support an effect of CD8+ cells in killing cells prior to viral integration. Sensitivity analyses of these results confirm that effect but also allow for additional effects, which the data do not discriminate well. Overall, we show that INT does not significantly increase the levels of 2-LTR circles. However, CD8+ cell depletion increases the 2-LTR levels, which are enhanced in the presence of an INT.IMPORTANCE CD8+ T cells play an essential role in controlling HIV and SIV infection, but the specific mechanisms involved remain poorly understood. Due to failed viral infection, HIV and SIV can form 2-LTR extrachromosomal circles that can be quantified. We present novel data on the dynamics of these 2-LTR forms in a SIV-infected macaque model under three different treatment conditions: depletion of CD8+ cells, administration of the integrase inhibitor in a monotherapy, which favors the formation of 2-LTR circles, and a combination of the two treatments. We used a new mathematical model to help interpret the data, and the results suggest that CD8+ cells exert a killing effect on infected cells prior to virus integration. These results provide new insights into the mechanisms of action of CD8+ cells in SIV infection. Confirmation of our results would be an important step in understanding immune control of HIV.
Collapse
|
39
|
Pinzone MR, O’Doherty U. Measuring integrated HIV DNA ex vivo and in vitro provides insights about how reservoirs are formed and maintained. Retrovirology 2018; 15:22. [PMID: 29452580 PMCID: PMC5816390 DOI: 10.1186/s12977-018-0396-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/19/2018] [Indexed: 11/29/2022] Open
Abstract
The identification of the most appropriate marker to measure reservoir size has been a great challenge for the HIV field. Quantitative viral outgrowth assay (QVOA), the reference standard to quantify the amount of replication-competent virus, has several limitations, as it is laborious, expensive, and unable to robustly reactivate every single integrated provirus. PCR-based assays have been developed as an easier, cheaper and less error-prone alternative to QVOA, but also have limitations. Historically, measuring integrated HIV DNA has provided insights about how reservoirs are formed and maintained. In the 1990s, measuring integrated HIV DNA was instrumental in understanding that a subset of resting CD4 T cells containing integrated HIV DNA were the major source of replication-competent virus. Follow-up studies have further characterized the phenotype of these cells containing integrated HIV DNA, as well as shown the correlation between the integration levels and clinical parameters, such as duration of infection, CD4 count and viral load. Integrated HIV DNA correlates with total HIV measures and with QVOA. The integration assay has several limitations. First, it largely overestimates the reservoir size, as both defective and replication-competent proviruses are detected. Since defective proviruses are the majority in patients on ART, it follows that the number of proviruses capable of reactivating and releasing new virions is significantly smaller than the number of integrated proviruses. Second, in patients on ART clonal expansion could theoretically lead to the preferential amplification of proviruses close to an Alu sequence though longitudinal studies have not captured this effect. Proviral sequencing combined with integration measures is probably the best estimate of reservoir size, but it is expensive, time-consuming and requires considerable bioinformatics expertise. All these reasons limit its use on a large scale. Herein, we review the utility of measuring HIV integration and suggest combining it with sequencing and total HIV measurements can provide insights that underlie reservoir maintenance.
Collapse
Affiliation(s)
- Marilia Rita Pinzone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Una O’Doherty
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|